1
|
El Cheikh J, Hamed F, Rifi H, Dakroub AH, Eid AH. Genetic polymorphisms influencing antihypertensive drug responses. Br J Pharmacol 2025; 182:929-950. [PMID: 39627167 DOI: 10.1111/bph.17414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 10/30/2024] [Accepted: 11/01/2024] [Indexed: 01/11/2025] Open
Abstract
Hypertension is a major contributor to cardiovascular disease and its associated morbidity and mortality. The low efficacy observed with some anti-hypertensive therapies has been attributed partly to inter-individual genetic variability. This paper reviews the major findings regarding these genetic variabilities that modulate responses to anti-hypertensive therapies such as angiotensin converting enzyme (ACE) inhibitors, angiotensin receptor blockers (ARBs), diuretics, calcium channel blockers (CCBs) and β-adrenoceptor blockers. The importance of studying these genetic polymorphisms stems from the goal to optimise anti-hypertensive therapy for each individual patient, aiming for the highest efficacy and lowest risk of adverse effects. It is important to recognise that environmental and epigenetic factors can contribute to the observed variations in drug responses. Owing to the multigenic and multifactorial nature of drug responses, further research is crucial for translating these findings into clinical practice and the establishment of reliable recommendations.
Collapse
Affiliation(s)
- Jana El Cheikh
- Faculty of Medicine, University of Balamand, Al Koura, Tripoli, Lebanon
| | - Fouad Hamed
- Faculty of Medicine, University of Balamand, Al Koura, Tripoli, Lebanon
| | - Hana Rifi
- Faculty of Medicine, University of Balamand, Al Koura, Tripoli, Lebanon
| | - Ali H Dakroub
- Blavatnik Family Research Institute, Departments of Cardiology and Population Health Science and Policy, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Ali Hussein Eid
- Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha, Qatar
| |
Collapse
|
2
|
Alghamdi MA, AL-Eitan L, Ibdah R, Bani Khalid I, Darabseh S, Alasmar M, Ataa A. Genomic Insights into Blood Pressure Regulation: Exploring Ion Channel and Transporter Gene Variations in Jordanian Hypertensive Individuals. MEDICINA (KAUNAS, LITHUANIA) 2025; 61:156. [PMID: 39859138 PMCID: PMC11766771 DOI: 10.3390/medicina61010156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 12/30/2024] [Accepted: 01/14/2025] [Indexed: 01/30/2025]
Abstract
Background and Objectives: Hypertension (HTN) constitutes a significant global health burden, yet the specific genetic variant responsible for blood pressure regulation remains elusive. This study investigates the genetic basis of hypertension in the Jordanian population, focusing on gene variants related to ion channels and transporters, including KCNJ1, WNK1, NPPA, STK39, LUC7L2, NEDD4L, NPHS1, BDKRB2, and CACNA1C. Materials and Methods: This research involved 200 hypertensive patients and 224 healthy controls. Whole blood samples were collected from each participant, and genomic DNA was extracted. The genetic distribution of the polymorphisms was analyzed. The haplotype frequencies were investigated using the SNPStats web tool, and the genotype and allele frequencies of the studied variants were assessed using the χ2 test. Results: Sixteen single nucleotide polymorphisms (SNPs) from nine genes were evaluated. A significant association was observed between the rs880054 variant of the WNK1 gene and hypertension susceptibility, with the T allele elevating the risk of hypertension. This association remained important in the codominant model (p = 0.049) and the dominant model (p = 0.029). In addition, rs880054 was associated with clinical characteristics such as triglyceride levels and cerebrovascular accidents (p-value > 0.05). Conclusions: Our findings reveal a significant link between the rs880054 SNP and an increased hypertension risk, suggesting that variations in WNK1 may be crucial in regulating blood pressure. This study provides new insights into the genetic factors contributing to hypertension and highlights the potential of WNK1 as a target for future therapeutic interventions.
Collapse
Affiliation(s)
- Mansour Abdullah Alghamdi
- Department of Anatomy, College of Medicine, King Khalid University, Abha 62529, Saudi Arabia
- Genomics and Personalized Medicine Unit, College of Medicine, King Khalid University, Abha 62529, Saudi Arabia
| | - Laith AL-Eitan
- Department of Biotechnology and Genetic Engineering, Jordan University of Science and Technology, Irbid 22110, Jordan (A.A.)
| | - Rasheed Ibdah
- Internal Medicine Department, College of Medicine, Jordan University of Science and Technology, Irbid 22110, Jordan
| | - Islam Bani Khalid
- Department of Biotechnology and Genetic Engineering, Jordan University of Science and Technology, Irbid 22110, Jordan (A.A.)
| | - Salma Darabseh
- Department of Biotechnology and Genetic Engineering, Jordan University of Science and Technology, Irbid 22110, Jordan (A.A.)
| | - Maryam Alasmar
- Department of Biotechnology and Genetic Engineering, Jordan University of Science and Technology, Irbid 22110, Jordan (A.A.)
| | - Asaad Ataa
- Department of Biotechnology and Genetic Engineering, Jordan University of Science and Technology, Irbid 22110, Jordan (A.A.)
| |
Collapse
|
3
|
Katsukunya JN, Jones E, Soko ND, Blom D, Sinxadi P, Rayner B, Dandara C. Genetic Variation in ABCB1, ADRB1, CYP3A4, CYP3A5, NEDD4L and NR3C2 Confers Differential Susceptibility to Resistant Hypertension among South Africans. J Pers Med 2024; 14:664. [PMID: 39063918 PMCID: PMC11277774 DOI: 10.3390/jpm14070664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 06/13/2024] [Accepted: 06/17/2024] [Indexed: 07/28/2024] Open
Abstract
Resistant hypertension (RHTN) prevalence ranges from 4 to 19% in Africa. There is a paucity of data on the role of genetic variation on RHTN among Africans. We set out to investigate the role of polymorphisms in ABCB1, ADRB1, CYP3A4, CYP3A5, NEDD4L, and NR3C2, on RHTN susceptibility among South Africans. Using a retrospective matched case-control study, 190 RHTN patients (cases: blood pressure (BP) ≥ 140/90 mmHg on ≥3 anti-hypertensives or BP < 140/90 mmHg on >3 anti-hypertensives) and 189 non-RHTN patients (controls: <3 anti-hypertensives, BP < 140/90 or ≥140/90 mmHg), 12 single nucleotide polymorphisms were genotyped using polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP), quantitative PCR and Sanger sequencing. Genetic association analyses were conducted using the additive model and multivariable logistic regression. Homozygosity for CYP3A5 rs776746C/C genotype (p = 0.02; OR: 0.44; CI: 0.22-0.89) was associated with reduced risk for RHTN. Homozygous ADRB1 rs1801252G/G (p = 0.02; OR: 3.30; CI: 1.17-10.03) and NEDD4L rs4149601A/A genotypes (p = 0.001; OR: 3.82; CI: 1.67-9.07) were associated with increased risk for RHTN. Carriers of the of ADRB1 rs1801252-rs1801253 G-C haplotype had 2.83-fold odds of presenting with RHTN (p = 0.04; OR: 2.83; CI: 1.05-8.20). These variants that are associated with RHTN may have clinical utility in the selection of antihypertensive drugs in our population.
Collapse
Affiliation(s)
- Jonathan N. Katsukunya
- Division of Human Genetics, Department of Pathology, Institute of Infectious Disease and Molecular Medicine (IDM), Faculty of Health Sciences, University of Cape Town, Cape Town 7700, South Africa; (J.N.K.); (N.D.S.)
- SAMRC/UCT Platform for Pharmacogenomics Research and Translation, South African Medical Research Council, Cape Town 7501, South Africa; (E.J.); (D.B.); (P.S.); (B.R.)
| | - Erika Jones
- SAMRC/UCT Platform for Pharmacogenomics Research and Translation, South African Medical Research Council, Cape Town 7501, South Africa; (E.J.); (D.B.); (P.S.); (B.R.)
- Department of Medicine, Division of Nephrology and Hypertension, Groote Schuur Hospital and Faculty of Health Sciences, University of Cape Town, Cape Town 7700, South Africa
| | - Nyarai D. Soko
- Division of Human Genetics, Department of Pathology, Institute of Infectious Disease and Molecular Medicine (IDM), Faculty of Health Sciences, University of Cape Town, Cape Town 7700, South Africa; (J.N.K.); (N.D.S.)
- SAMRC/UCT Platform for Pharmacogenomics Research and Translation, South African Medical Research Council, Cape Town 7501, South Africa; (E.J.); (D.B.); (P.S.); (B.R.)
- Department of Pharmaceutical Technology, School of Allied Health Sciences, Harare Institute of Technology, Harare P.O. Box BE 277, Zimbabwe
| | - Dirk Blom
- SAMRC/UCT Platform for Pharmacogenomics Research and Translation, South African Medical Research Council, Cape Town 7501, South Africa; (E.J.); (D.B.); (P.S.); (B.R.)
- Department of Medicine, Division of Lipidology and Cape Heart Institute, Groote Schuur Hospital and Faculty of Health Sciences, University of Cape Town, Cape Town 7700, South Africa
| | - Phumla Sinxadi
- SAMRC/UCT Platform for Pharmacogenomics Research and Translation, South African Medical Research Council, Cape Town 7501, South Africa; (E.J.); (D.B.); (P.S.); (B.R.)
- Department of Medicine, Division of Clinical Pharmacology, Groote Schuur Hospital and Faculty of Health Sciences, University of Cape Town, Cape Town 7700, South Africa
| | - Brian Rayner
- SAMRC/UCT Platform for Pharmacogenomics Research and Translation, South African Medical Research Council, Cape Town 7501, South Africa; (E.J.); (D.B.); (P.S.); (B.R.)
- Department of Medicine, Division of Nephrology and Hypertension, Groote Schuur Hospital and Faculty of Health Sciences, University of Cape Town, Cape Town 7700, South Africa
| | - Collet Dandara
- Division of Human Genetics, Department of Pathology, Institute of Infectious Disease and Molecular Medicine (IDM), Faculty of Health Sciences, University of Cape Town, Cape Town 7700, South Africa; (J.N.K.); (N.D.S.)
- SAMRC/UCT Platform for Pharmacogenomics Research and Translation, South African Medical Research Council, Cape Town 7501, South Africa; (E.J.); (D.B.); (P.S.); (B.R.)
| |
Collapse
|
4
|
You JR, Wen ZJ, Tian JW, Lv XB, Li R, Li SP, Xin H, Li PF, Zhang YF, Zhang R. Crosstalk between ubiquitin ligases and ncRNAs drives cardiovascular disease progression. Front Immunol 2024; 15:1335519. [PMID: 38515760 PMCID: PMC10954775 DOI: 10.3389/fimmu.2024.1335519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 02/26/2024] [Indexed: 03/23/2024] Open
Abstract
Cardiovascular diseases (CVDs) are multifactorial chronic diseases and have the highest rates of morbidity and mortality worldwide. The ubiquitin-proteasome system (UPS) plays a crucial role in posttranslational modification and quality control of proteins, maintaining intracellular homeostasis via degradation of misfolded, short-lived, or nonfunctional regulatory proteins. Noncoding RNAs (ncRNAs, such as microRNAs, long noncoding RNAs, circular RNAs and small interfering RNAs) serve as epigenetic factors and directly or indirectly participate in various physiological and pathological processes. NcRNAs that regulate ubiquitination or are regulated by the UPS are involved in the execution of target protein stability. The cross-linked relationship between the UPS, ncRNAs and CVDs has drawn researchers' attention. Herein, we provide an update on recent developments and perspectives on how the crosstalk of the UPS and ncRNAs affects the pathological mechanisms of CVDs, particularly myocardial ischemia/reperfusion injury, myocardial infarction, cardiomyopathy, heart failure, atherosclerosis, hypertension, and ischemic stroke. In addition, we further envision that RNA interference or ncRNA mimics or inhibitors targeting the UPS can potentially be used as therapeutic tools and strategies.
Collapse
Affiliation(s)
- Jia-Rui You
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, Shandong, China
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, China
| | - Zeng-Jin Wen
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, Shandong, China
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, China
| | - Jia-Wei Tian
- Department of Emergency Internal Medicine, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Xiao-Bing Lv
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, Shandong, China
| | - Rong Li
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, Shandong, China
| | - Shu-Ping Li
- Department of Cardiology, The Affiliated Qingdao Third People’s Hospital of Qingdao University, Qingdao University, Qingdao, Shandong, China
| | - Hui Xin
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, Shandong, China
| | - Pei-Feng Li
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, China
| | - Yin-Feng Zhang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, China
| | - Rui Zhang
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, Shandong, China
| |
Collapse
|
5
|
Katsukunya JN, Soko ND, Naidoo J, Rayner B, Blom D, Sinxadi P, Chimusa ER, Dandara M, Dzobo K, Jones E, Dandara C. Pharmacogenomics of Hypertension in Africa: Paving the Way for a Pharmacogenetic-Based Approach for the Treatment of Hypertension in Africans. Int J Hypertens 2023; 2023:9919677. [PMID: 38633331 PMCID: PMC11022520 DOI: 10.1155/2023/9919677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 04/21/2023] [Accepted: 05/22/2023] [Indexed: 04/19/2024] Open
Abstract
In Africa, the burden of hypertension has been rising at an alarming rate for the last two decades and is a major cause for cardiovascular disease (CVD) mortality and morbidity. Hypertension is characterised by elevated blood pressure (BP) ≥ 140/90 mmHg. Current hypertension guidelines recommend the use of antihypertensives belonging to the following classes: calcium channel blockers (CCB), angiotensin converting inhibitors (ACEI), angiotensin receptor blockers (ARB), diuretics, β-blockers, and mineralocorticoid receptor antagonists (MRAs), to manage hypertension. Still, a considerable number of hypertensives in Africa have their BP uncontrolled due to poor drug response and remain at the risk of CVD events. Genetic factors are a major contributing factor, accounting for 20% to 80% of individual variability in therapy and poor response. Poor response to antihypertensive drug therapy is characterised by elevated BPs and occurrence of adverse drug reactions (ADRs). As a result, there have been numerous studies which have examined the role of genetic variation and its influence on antihypertensive drug response. These studies are predominantly carried out in non-African populations, including Europeans and Asians, with few or no Africans participating. It is important to note that the greatest genetic diversity is observed in African populations as well as the highest prevalence of hypertension. As a result, this warrants a need to focus on how genetic variation affects response to therapeutic interventions used to manage hypertension in African populations. In this paper, we discuss the implications of genetic diversity in CYP11B2, GRK4, NEDD4L, NPPA, SCNN1B, UMOD, CYP411, WNK, CYP3A4/5, ACE, ADBR1/2, GNB3, NOS3, B2, BEST3, SLC25A31, LRRC15 genes, and chromosome 12q loci on hypertension susceptibility and response to antihypertensive therapy. We show that African populations are poorly explored genetically, and for the few characterised genes, they exhibit qualitative and quantitative differences in the profile of pharmacogene variants when compared to other ethnic groups. We conclude by proposing prioritization of pharmacogenetics research in Africa and possible adoption of pharmacogenetic-guided therapies for hypertension in African patients. Finally, we outline the implications, challenges, and opportunities these studies present for populations of non-European descent.
Collapse
Affiliation(s)
- Jonathan N. Katsukunya
- Division of Human Genetics, Department of Pathology and Institute of Infectious Disease and Molecular Medicine (IDM), Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
- UCT/South African Medical Research Council (SAMRC) Platform for Pharmacogenomics Research and Translation Unit, University of Cape Town, Cape Town, South Africa
| | - Nyarai D. Soko
- Division of Human Genetics, Department of Pathology and Institute of Infectious Disease and Molecular Medicine (IDM), Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
- UCT/South African Medical Research Council (SAMRC) Platform for Pharmacogenomics Research and Translation Unit, University of Cape Town, Cape Town, South Africa
| | - Jashira Naidoo
- Department of Medicine, Division of Nephrology and Hypertension, Groote Schuur Hospital and Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Brian Rayner
- UCT/South African Medical Research Council (SAMRC) Platform for Pharmacogenomics Research and Translation Unit, University of Cape Town, Cape Town, South Africa
- Department of Medicine, Division of Nephrology and Hypertension, Groote Schuur Hospital and Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Dirk Blom
- UCT/South African Medical Research Council (SAMRC) Platform for Pharmacogenomics Research and Translation Unit, University of Cape Town, Cape Town, South Africa
- Department of Medicine, Division of Lipidology and Cape Heart Institute, Groote Schuur Hospital and Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Phumla Sinxadi
- UCT/South African Medical Research Council (SAMRC) Platform for Pharmacogenomics Research and Translation Unit, University of Cape Town, Cape Town, South Africa
- Department of Medicine, Division of Clinical Pharmacology, Groote Schuur Hospital and Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Emile R. Chimusa
- Department of Applied Sciences, Faculty of Health and Life Sciences, Northumbria University, Newcastle, Tyne and Wear NE1 8ST, UK
| | - Michelle Dandara
- UCT/South African Medical Research Council (SAMRC) Platform for Pharmacogenomics Research and Translation Unit, University of Cape Town, Cape Town, South Africa
| | - Kevin Dzobo
- Medical Research Council-SA Wound Healing Unit, Hair and Skin Research Laboratory, Division of Dermatology, Department of Medicine, Groote Schuur Hospital, Faculty of Health Sciences University of Cape Town, Anzio Road Observatory, Cape Town 7925, South Africa
| | - Erika Jones
- UCT/South African Medical Research Council (SAMRC) Platform for Pharmacogenomics Research and Translation Unit, University of Cape Town, Cape Town, South Africa
- Department of Medicine, Division of Nephrology and Hypertension, Groote Schuur Hospital and Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Collet Dandara
- Division of Human Genetics, Department of Pathology and Institute of Infectious Disease and Molecular Medicine (IDM), Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
- UCT/South African Medical Research Council (SAMRC) Platform for Pharmacogenomics Research and Translation Unit, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
6
|
Chen Y, Han Y, Wu Y, Hui R, Yang Y, Zhong Y, Zhang S, Zhang W. Pharmacogenetic association of the NR1H3 promoter variant with antihypertensive response among patients with hypertension: A longitudinal study. Front Pharmacol 2023; 14:1083134. [PMID: 36950018 PMCID: PMC10025344 DOI: 10.3389/fphar.2023.1083134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Accepted: 02/20/2023] [Indexed: 03/08/2023] Open
Abstract
Background: The genetic factors in assessing therapeutic efficacy and predicting antihypertensive drug response are unclear. Therefore, this study aims to identify the associations between variants and antihypertensive drug response. Methods: A longitudinal study including 1837 hypertensive patients was conducted in Northern China and followed up for a median 2.24 years. The associations of 11 candidate variants with blood pressure changes in response to antihypertensive drugs and with the risk of cardiovascular events during the follow-up were examined. The dual-luciferase assay was carried out to assess the effect of genetic variants on gene transcriptional activity. Results: The variant rs11039149A>G in the promoter of nuclear receptor subfamily 1 group H member 3 (NR1H3) was associated with the change in systolic blood pressure (ΔSBP) in response to calcium channel blockers (CCBs) monotherapy. Patients carrying rs11039149AG genotype showed a significant increase of systolic blood pressure (SBP) at follow-up compared with AA carriers, and the difference of ΔSBP between AG and AA carriers was 5.94 mm Hg (95%CI: 2.09-9.78, p = 0.002). In 1,184 patients with CCBs therapy, SBP levels decreased in AA carriers, but increased in AG carriers, the difference of ΔSBP between AG and AA carriers was 8.04 mm Hg (95%CI: 3.28-12.81, p = 0.001). Further analysis in 359 patients with CCBs monotherapy, the difference of ΔSBP between AG and AA carriers was 15.25 mm Hg (95%CI: 6.48-24.02, p = 0.001). However, there was no significant difference in ΔSBP between AG and AA carriers with CCBs multitherapy. The rs11039149A>G was not associated with the cardiovascular events incidence during the follow-up. Additionally, transcriptional factor forkhead box C1 (FOXC1) bound to the NR1H3 promoter containing rs11039149A and significantly increased the transcriptional activity, while rs11039149 A to G change led to a loss-of-function and disabled FOXC1 binding. For the other 10 variants, associations with blood pressure changes or risk of cardiovascular events were not observed. Conclusion: Hypertensive patients with rs11039149AG genotype in the NR1H3 gene have a significant worse SBP control in response to CCBs monotherapy compared with AA carriers. Our findings suggest that the NR1H3 gene might act as a promising genetic factor to affect individual sensitivity to antihypertensive drugs.
Collapse
Affiliation(s)
- Yu Chen
- National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, National Center for Cardiovascular Diseases, Beijing, China
| | - Yuqing Han
- National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, National Center for Cardiovascular Diseases, Beijing, China
| | - Yiyi Wu
- The First Affiliated Hospital of Anhui University of Science and Technology, The First People’s Hospital of Huainan City, Huainan, Anhui, China
| | - Rutai Hui
- National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, National Center for Cardiovascular Diseases, Beijing, China
| | - Yunyun Yang
- Clinical Laboratory, Xiamen Key Laboratory of Genetic Testing, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian, China
| | - Yixuan Zhong
- National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, National Center for Cardiovascular Diseases, Beijing, China
| | - Shuyuan Zhang
- National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, National Center for Cardiovascular Diseases, Beijing, China
| | - Weili Zhang
- National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, National Center for Cardiovascular Diseases, Beijing, China
- Central-China Branch of National Center for Cardiovascular Diseases, Henan Cardiovascular Disease Center, Fuwai Central-China Hospital, Zhengzhou, China
- *Correspondence: Weili Zhang,
| |
Collapse
|
7
|
Dong M, Liu J, Liu C, Wang H, Sun W, Liu B. CRISPR/CAS9: A promising approach for the research and treatment of cardiovascular diseases. Pharmacol Res 2022; 185:106480. [PMID: 36191879 DOI: 10.1016/j.phrs.2022.106480] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Revised: 09/27/2022] [Accepted: 09/29/2022] [Indexed: 10/31/2022]
Abstract
The development of gene-editing technology has been one of the biggest advances in biomedicine over the past two decades. Not only can it be used as a research tool to build a variety of disease models for the exploration of disease pathogenesis at the genetic level, it can also be used for prevention and treatment. This is done by intervening with the expression of target genes and carrying out precise molecular targeted therapy for diseases. The simple and flexible clustered regularly interspaced short palindromic repeats (CRISPR)/Cas9 gene-editing technology overcomes the limitations of zinc finger nucleases (ZFNs) and transcription activator-like effector nucleases (TALENs). For this reason, it has rapidly become a preferred method for gene editing. As a new gene intervention method, CRISPR/Cas9 has been widely used in the clinical treatment of tumours and rare diseases; however, its application in the field of cardiovascular diseases is currently limited. This article reviews the application of the CRISPR/Cas9 editing technology in cardiovascular disease research and treatment, and discusses the limitations and prospects of this technology.
Collapse
Affiliation(s)
- Mengying Dong
- Department of Cardiology, The Second Hospital of Jilin University, 218 Ziqiang Road, Changchun, China, 130041
| | - Jiangen Liu
- Department of Cardiology, The Second Hospital of Jilin University, 218 Ziqiang Road, Changchun, China, 130041
| | - Caixia Liu
- Department of Neurology, The Liaoning Province People's Hospital, 33 Wenyi Road, ShenYang, China, 110016
| | - He Wang
- Department of Cardiology, The Second Hospital of Jilin University, 218 Ziqiang Road, Changchun, China, 130041
| | - Wei Sun
- Department of Cardiology, The Second Hospital of Jilin University, 218 Ziqiang Road, Changchun, China, 130041.
| | - Bin Liu
- Department of Cardiology, The Second Hospital of Jilin University, 218 Ziqiang Road, Changchun, China, 130041.
| |
Collapse
|
8
|
Jin X, Zhang L, Ji J, Ju T, Zhao J, Yuan Z. Network regression analysis in transcriptome-wide association studies. BMC Genomics 2022; 23:562. [PMID: 35933330 PMCID: PMC9356418 DOI: 10.1186/s12864-022-08809-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 08/02/2022] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Transcriptome-wide association studies (TWASs) have shown great promise in interpreting the findings from genome-wide association studies (GWASs) and exploring the disease mechanisms, by integrating GWAS and eQTL mapping studies. Almost all TWAS methods only focus on one gene at a time, with exception of only two published multiple-gene methods nevertheless failing to account for the inter-dependence as well as the network structure among multiple genes, which may lead to power loss in TWAS analysis as complex disease often owe to multiple genes that interact with each other as a biological network. We therefore developed a Network Regression method in a two-stage TWAS framework (NeRiT) to detect whether a given network is associated with the traits of interest. NeRiT adopts the flexible Bayesian Dirichlet process regression to obtain the gene expression prediction weights in the first stage, uses pointwise mutual information to represent the general between-node correlation in the second stage and can effectively take the network structure among different gene nodes into account. RESULTS Comprehensive and realistic simulations indicated NeRiT had calibrated type I error control for testing both the node effect and edge effect, and yields higher power than the existed methods, especially in testing the edge effect. The results were consistent regardless of the GWAS sample size, the gene expression prediction model in the first step of TWAS, the network structure as well as the correlation pattern among different gene nodes. Real data applications through analyzing systolic blood pressure and diastolic blood pressure from UK Biobank showed that NeRiT can simultaneously identify the trait-related nodes as well as the trait-related edges. CONCLUSIONS NeRiT is a powerful and efficient network regression method in TWAS.
Collapse
Affiliation(s)
- Xiuyuan Jin
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China.,Institute for Medical Dataology, Shandong University, Jinan, 250003, Shandong, China
| | - Liye Zhang
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China.,Institute for Medical Dataology, Shandong University, Jinan, 250003, Shandong, China
| | - Jiadong Ji
- Institute for Financial Studies, Shandong University, Jinan, 250100, Shandong, China
| | - Tao Ju
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China.,Institute for Medical Dataology, Shandong University, Jinan, 250003, Shandong, China
| | - Jinghua Zhao
- Department of Public Health and Primary Care, Cardiovascular Epidemiology Unit, University of Cambridge, Cambridge, UK.
| | - Zhongshang Yuan
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China. .,Institute for Medical Dataology, Shandong University, Jinan, 250003, Shandong, China.
| |
Collapse
|
9
|
Abstract
Hypertension is a frequent finding in elderly patients. Hypertension in older age can be both associated with frailty and represent a risk factor for frailty. Hypertension is recognized as a main risk factor for cardiovascular diseases such as heart failure, atrial fibrillation, and stroke and the occurrence of these diseases may provoke a decline in health status and/or worsen the degree of frailty. Blood pressure targets in hypertensive older and frail patients are not completely defined. However, specific evaluations of individual patients and their co-morbidities and assessment of domains and components of frailty, together with weighted consideration of drug use, may help in finding the appropriate therapy.
Collapse
|
10
|
Khan AR, Shah SH, Ajaz S, Firasat S, Abid A, Raza A. The Prevalence of Pharmacogenomics Variants and Their Clinical Relevance Among the Pakistani Population. Evol Bioinform Online 2022; 18:11769343221095834. [PMID: 35497687 PMCID: PMC9047794 DOI: 10.1177/11769343221095834] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Accepted: 04/04/2022] [Indexed: 11/28/2022] Open
Abstract
Background: Pharmacogenomics (PGx), forming the basis of precision medicine, has
revolutionized traditional medical practice. Currently, drug responses such
as drug efficacy, drug dosage, and drug adverse reactions can be anticipated
based on the genetic makeup of the patients. The pharmacogenomic data of
Pakistani populations are limited. This study investigates the frequencies
of pharmacogenetic variants and their clinical relevance among ethnic groups
in Pakistan. Methods: The Pharmacogenomics Knowledge Base (PharmGKB) database was used to extract
pharmacogenetic variants that are involved in medical conditions with high
(1A + 1B) to moderate (2A + 2B) clinical evidence. Subsequently, the allele
frequencies of these variants were searched among multiethnic groups of
Pakistan (Balochi, Brahui, Burusho, Hazara, Kalash, Pashtun, Punjabi, and
Sindhi) using the 1000 Genomes Project (1KGP) and
ALlele FREquency
Database (ALFRED). Furthermore, the published
Pharmacogenomics literature on the Pakistani population was reviewed in
PubMed and Google Scholar. Results: Our search retrieved (n = 29) pharmacogenetic genes and their (n = 44)
variants with high to moderate evidence of clinical association. These
pharmacogenetic variants correspond to drug-metabolizing enzymes (n = 22),
drug-metabolizing transporters (n = 8), and PGx gene regulators, etc.
(n = 14). We found 5 pharmacogenetic variants present at >50% among 8
ethnic groups of Pakistan. These pharmacogenetic variants include
CYP2B6 (rs2279345, C; 70%-86%), CYP3A5
(rs776746, C; 64%-88%), FLT3 (rs1933437, T; 54%-74%),
CETP (rs1532624, A; 50%-70%), and DPP6
(rs6977820, C; 61%-86%) genes that are involved in drug response for
acquired immune deficiency syndrome, transplantation, cancer, heart disease,
and mental health therapy, respectively. Conclusions: This study highlights the frequency of important clinical pharmacogenetic
variants (1A, 1B, 2A, and 2B) among multi-ethnic Pakistani populations. The
high prevalence (>50%) of single nucleotide pharmacogenetic variants may
contribute to the drug response/diseases outcome. These PGx data could be
used as pharmacogenetic markers in the selection of appropriate therapeutic
regimens for specific ethnic groups of Pakistan.
Collapse
Affiliation(s)
- Abdul Rafay Khan
- Center for Human Genetics and Molecular Medicine, Sindh Institute of Urology and Transplantation, Karachi, Pakistan
| | - Sayed Hajan Shah
- Center for Human Genetics and Molecular Medicine, Sindh Institute of Urology and Transplantation, Karachi, Pakistan
| | - Sadia Ajaz
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, Pakistan
| | - Sadaf Firasat
- Center for Human Genetics and Molecular Medicine, Sindh Institute of Urology and Transplantation, Karachi, Pakistan
| | - Aiysha Abid
- Center for Human Genetics and Molecular Medicine, Sindh Institute of Urology and Transplantation, Karachi, Pakistan
| | - Ali Raza
- Center for Human Genetics and Molecular Medicine, Sindh Institute of Urology and Transplantation, Karachi, Pakistan
| |
Collapse
|
11
|
Shi Y, Li X, Yang J. Cx43 upregulation in HUVECs under stretch via TGF-β1 and cytoskeletal network. Open Med (Wars) 2022; 17:463-474. [PMID: 35350835 PMCID: PMC8919824 DOI: 10.1515/med-2022-0432] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 01/11/2022] [Accepted: 01/14/2022] [Indexed: 12/25/2022] Open
Abstract
Many physiological and pathophysiological processes in cells or tissues are involved in mechanical stretch, which induces the gap junction gene expression and cytokine TGF beta changes. However, the underlying mechanisms of the gap junction gene expression remain unknown. Here, we showed that the mRNA and protein levels of Cx43 in human umbilical vein endothelial cells (HUVECs) were significantly increased after 24 h stretch stimulation, and TGF beta1 (not TGF beta2) expression was also upregulated. Administration of TGF beta1 into HUVECs without stretch also induced upregulation of Cx43 expression. However, SB431542, a specific inhibitor of the TGF beta1 receptor, blocked the Cx43 protein upregulation caused by TGF beta1. Further, the increase of Cx43 protein expression under the stretch condition was partially blocked by SB431542; it was also partially blocked by simultaneous administration of anti-TGF beta1 monoclonal neutralization antibody. Importantly, the upregulation of Cx43 induced by stretch was blocked by the administration of actin and microtubule inhibitors, while NEDD4, a key element in mediating Cx43 protein ubiquitination and degradation, was not changed under the stretch condition. In conclusion, upregulation of Cx43 expression under the 24 h stretch condition is mediated via TGF beta1 receptor signaling pathway, and it also involves the actin and microtubule cytoskeletal network.
Collapse
Affiliation(s)
- Yumeng Shi
- Department of Ophthalmology and Visual Science, Eye Ear Nose and Throat Hospital of Fudan University, Shanghai 200031, China
| | - Xinbo Li
- Department of Ophthalmology, Casey Eye Institute, Oregon Health & Science University, Oregon, USA
| | - Jin Yang
- Department of Ophthalmology and Visual Science, Eye Ear Nose and Throat Hospital of Fudan University, Shanghai 200031, China
| |
Collapse
|
12
|
Eadon MT, Maddatu J, Moe SM, Sinha AD, Melo Ferreira R, Miller BW, Sher SJ, Su J, Pratt VM, Chapman AB, Skaar TC, Moorthi RN. Pharmacogenomics of Hypertension in CKD: The CKD-PGX Study. KIDNEY360 2022; 3:307-316. [PMID: 35342886 PMCID: PMC8953763 DOI: 10.34067/kid.0005362021] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 12/08/2021] [Indexed: 01/12/2023]
Abstract
Background Patients with CKD often have uncontrolled hypertension despite polypharmacy. Pharmacogenomic drug-gene interactions (DGIs) may affect the metabolism or efficacy of antihypertensive agents. We report changes in hypertension control after providing a panel of 11 pharmacogenomic predictors of antihypertensive response. Methods A prospective cohort with CKD and hypertension was followed to assess feasibility of pharmacogenomic testing implementation, self-reported provider utilization, and BP control. The analysis population included 382 subjects with hypertension who were genotyped for cross-sectional assessment of DGIs, and 335 subjects followed for 1 year to assess systolic BP (SBP) and diastolic BP (DBP). Results Most participants (58%) with uncontrolled hypertension had a DGI reducing the efficacy of one or more antihypertensive agents. Subjects with a DGI had 1.85-fold (95% CI, 1.2- to 2.8-fold) higher odds of uncontrolled hypertension, as compared with those without a DGI, adjusted for race, health system (safety-net hospital versus other locations), and advanced CKD (eGFR <30 ml/min). CYP2C9-reduced metabolism genotypes were associated with losartan response and uncontrolled hypertension (odds ratio [OR], 5.2; 95% CI, 1.9 to 14.7). CYP2D6-intermediate or -poor metabolizers had less frequent uncontrolled hypertension compared with normal metabolizers taking metoprolol or carvedilol (OR, 0.55; 95% CI, 0.3 to 0.95). In 335 subjects completing 1-year follow-up, SBP (-4.0 mm Hg; 95% CI, 1.6 to 6.5 mm Hg) and DBP (-3.3 mm Hg; 95% CI, 2.0 to 4.6 mm Hg) were improved. No significant difference in SBP or DBP change were found between individuals with and without a DGI. Conclusions There is a potential role for the addition of pharmacogenomic testing to optimize antihypertensive regimens in patients with CKD.
Collapse
Affiliation(s)
- Michael T. Eadon
- Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana
- Richard L. Roudebush Veterans Administration Medical Center, Indianapolis, Indiana
| | - Judith Maddatu
- Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana
| | - Sharon M. Moe
- Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana
| | - Arjun D. Sinha
- Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana
- Richard L. Roudebush Veterans Administration Medical Center, Indianapolis, Indiana
| | - Ricardo Melo Ferreira
- Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana
| | - Brent W. Miller
- Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana
| | - S. Jawad Sher
- Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana
| | - Jing Su
- Department of Biostatistics and Health Data Science, Indiana University School of Medicine, Indianapolis, Indiana
| | - Victoria M. Pratt
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana
| | | | - Todd C. Skaar
- Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana
| | - Ranjani N. Moorthi
- Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana
| |
Collapse
|
13
|
Babayeva M, Azzi B, Loewy ZG. Pharmacogenomics Informs Cardiovascular Pharmacotherapy. Methods Mol Biol 2022; 2547:201-240. [PMID: 36068466 DOI: 10.1007/978-1-0716-2573-6_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Precision medicine exemplifies the emergence of personalized treatment options which may benefit specific patient populations based upon their genetic makeup. Application of pharmacogenomics requires an understanding of how genetic variations impact pharmacokinetic and pharmacodynamic properties. This particular approach in pharmacotherapy is helpful because it can assist in and improve clinical decisions. Application of pharmacogenomics to cardiovascular pharmacotherapy provides for the ability of the medical provider to gain critical knowledge on a patient's response to various treatment options and risk of side effects.
Collapse
Affiliation(s)
| | | | - Zvi G Loewy
- Touro College of Pharmacy, New York, NY, USA.
- School of Medicine, New York Medical College, Valhalla, NY, USA.
| |
Collapse
|
14
|
McDonough CW, Warren HR, Jack JR, Motsinger-Reif AA, Armstrong ND, Bis JC, House JS, Singh S, El Rouby NM, Gong Y, Mychaleckyj JC, Rotroff DM, Benavente OR, Caulfield MJ, Doria A, Pepine CJ, Psaty BM, Glorioso V, Glorioso N, Hiltunen TP, Kontula KK, Arnett DK, Buse JB, Irvin MR, Johnson JA, Munroe PB, Wagner MJ, Cooper-DeHoff RM. Adverse Cardiovascular Outcomes and Antihypertensive Treatment: A Genome-Wide Interaction Meta-Analysis in the International Consortium for Antihypertensive Pharmacogenomics Studies. Clin Pharmacol Ther 2021; 110:723-732. [PMID: 34231218 PMCID: PMC8672325 DOI: 10.1002/cpt.2355] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2021] [Accepted: 06/11/2021] [Indexed: 01/01/2023]
Abstract
We sought to identify genome-wide variants influencing antihypertensive drug response and adverse cardiovascular outcomes, utilizing data from four randomized controlled trials in the International Consortium for Antihypertensive Pharmacogenomics Studies (ICAPS). Genome-wide antihypertensive drug-single nucleotide polymorphism (SNP) interaction tests for four drug classes (β-blockers, n = 9,195; calcium channel blockers (CCBs), n = 10,511; thiazide/thiazide-like diuretics, n = 3,516; ACE-inhibitors/ARBs, n = 2,559) and cardiovascular outcomes (incident myocardial infarction, stroke, or death) were analyzed among patients with hypertension of European ancestry. Top SNPs from the meta-analyses were tested for replication of cardiovascular outcomes in an independent Cohorts for Heart and Aging Research in Genomic Epidemiology (CHARGE) study (n = 21,267), blood pressure (BP) response in independent ICAPS studies (n = 1,552), and ethnic validation in African Americans from the Genetics of Hypertension Associated Treatment study (GenHAT; n = 5,115). One signal reached genome-wide significance in the β-blocker-SNP interaction analysis (rs139945292, Interaction P = 1.56 × 10-8 ). rs139945292 was validated through BP response to β-blockers, with the T-allele associated with less BP reduction (systolic BP response P = 6 × 10-4 , Beta = 3.09, diastolic BP response P = 5 × 10-3 , Beta = 1.53). The T-allele was also associated with increased adverse cardiovascular risk within the β-blocker treated patients' subgroup (P = 2.35 × 10-4 , odds ratio = 1.57, 95% confidence interval = 1.23-1.99). The locus showed nominal replication in CHARGE, and consistent directional trends in β-blocker treated African Americans. rs139945292 is an expression quantitative trait locus for the 50 kb upstream gene NTM (neurotrimin). No SNPs attained genome-wide significance for any other drugs classes. Top SNPs were located near CALB1 (CCB), FLJ367777 (ACE-inhibitor), and CES5AP1 (thiazide). The NTM region is associated with increased risk for adverse cardiovascular outcomes and less BP reduction in β-blocker treated patients. Further investigation into this region is warranted.
Collapse
Affiliation(s)
- Caitrin W. McDonough
- Department of Pharmacotherapy and Translational Research and Center for Pharmacogenomics and Precision Medicine, College of Pharmacy, University of Florida, Gainesville, Florida, USA
| | - Helen R. Warren
- Clinical Pharmacology Department, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
- NIHR Cardiovascular Biomedical Research Centre, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - John R. Jack
- Bioinformatics Research Center, Department of Statistics, North Carolina State University, Raleigh, North Carolina, USA
| | - Alison A. Motsinger-Reif
- Biostatistics and Computational Biology Branch, National Institute of Environmental Health Sciences, Durham, North Carolina, USA
| | - Nicole D. Armstrong
- Department of Epidemiology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Joshua C. Bis
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, Washington, USA
| | - John S. House
- Biostatistics and Computational Biology Branch, National Institute of Environmental Health Sciences, Durham, North Carolina, USA
| | - Sonal Singh
- Department of Pharmacotherapy and Translational Research and Center for Pharmacogenomics and Precision Medicine, College of Pharmacy, University of Florida, Gainesville, Florida, USA
| | - Nihal M. El Rouby
- Department of Pharmacotherapy and Translational Research and Center for Pharmacogenomics and Precision Medicine, College of Pharmacy, University of Florida, Gainesville, Florida, USA
| | - Yan Gong
- Department of Pharmacotherapy and Translational Research and Center for Pharmacogenomics and Precision Medicine, College of Pharmacy, University of Florida, Gainesville, Florida, USA
| | - Joesyf C. Mychaleckyj
- Center for Public Health Genomics, University of Virginia, Charlottesville, Virginia, USA
| | - Daniel M. Rotroff
- Bioinformatics Research Center, Department of Statistics, North Carolina State University, Raleigh, North Carolina, USA
| | - Oscar R. Benavente
- Department of Neurology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Mark J. Caulfield
- National Institute for Health Research, Barts Cardiovascular Biomedical Research Center, Queen Mary University of London, London, UK
| | - Alessandrio Doria
- Research Division, Joslin Diabetes Center; and Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Carl J. Pepine
- Division of Cardiovascular Medicine, Department of Medicine, College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Bruce M. Psaty
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, Washington, USA
- Department of Epidemiology, University of Washington, Seattle, Washington, USA
| | - Valeria Glorioso
- Department of Statistics and Quantitative Methods, University of Milano-Bicocca, Milano, Italy
| | - Nicola Glorioso
- Department of Clinical, Surgical and Experimental Science, University of Sassari, Medical School, Sassari, Italy
| | - Timo P. Hiltunen
- Department of Medicine and Research Program for Clinical and Molecular Metabolism, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Kimmo K. Kontula
- Department of Medicine and Research Program for Clinical and Molecular Metabolism, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Donna K. Arnett
- College of Public Health, Dean’s Office, University of Kentucky, Lexington, Kentucky, USA
| | - John B. Buse
- Division of Endocrinology, Department of Medicine, UNC School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Marguerite R. Irvin
- Department of Epidemiology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Julie A. Johnson
- Department of Pharmacotherapy and Translational Research and Center for Pharmacogenomics and Precision Medicine, College of Pharmacy, University of Florida, Gainesville, Florida, USA
- Division of Cardiovascular Medicine, Department of Medicine, College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Patricia B. Munroe
- Clinical Pharmacology Department, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
- NIHR Cardiovascular Biomedical Research Centre, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Michael J. Wagner
- Center for Pharmacogenomics and Individualized Therapy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Rhonda M. Cooper-DeHoff
- Department of Pharmacotherapy and Translational Research and Center for Pharmacogenomics and Precision Medicine, College of Pharmacy, University of Florida, Gainesville, Florida, USA
- Division of Cardiovascular Medicine, Department of Medicine, College of Medicine, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
15
|
Rahman F, Muthaiah N, Kumaramanickavel G. Current concepts and molecular mechanisms in pharmacogenetics of essential hypertension. Indian J Pharmacol 2021; 53:301-309. [PMID: 34414909 PMCID: PMC8411967 DOI: 10.4103/ijp.ijp_593_19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Hypertension is a leading age-related disease in our society and if left untreated, leads to fatal cardiovascular complications. The prevalence of hypertension has increased and becomes a significant global health economic burden, particularly in lower-income societies. Many loci associated with blood pressure and hypertension have been reported by genome-wide association studies that provided potential targets for pharmacotherapy. Pharmacogenetic research had shown interindividual variations in drug efficacy, safety, and tolerability. This could be due to genetic polymorphisms in the pharmacokinetics (genes involved in a transporter, plasma protein binding, and metabolism) or pharmacodynamic pathway (receptors, ion channels, enzymes). Pharmacogenetics promises great hope toward targeted therapy, but challenges remain in implementing pharmacogenetic aided antihypertensive therapy in clinical practice. Using various databases, we analyzed the underlying mechanisms between the candidate gene polymorphisms and antihypertensive drug interactions and the challenges of implementing precision medicine. We review the emergence of pharmacogenetics and its relevance to clinical pharmacological practice.
Collapse
Affiliation(s)
- Farhana Rahman
- Department of Pharmacology, Sree Balaji Medical College and Hospital, Bharat University, Chennai, Tamil Nadu, India
| | - Nagasundaram Muthaiah
- Department of Pharmacology, Sree Balaji Medical College and Hospital, Bharat University, Chennai, Tamil Nadu, India
| | - Govindasamy Kumaramanickavel
- Genomic Research Centre, Sree Balaji Medical College and Hospital, Bharat University, Chennai, Tamil Nadu, India
| |
Collapse
|
16
|
McDonough CW. Pharmacogenomics in Cardiovascular Diseases. Curr Protoc 2021; 1:e189. [PMID: 34232575 DOI: 10.1002/cpz1.189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Cardiovascular pharmacogenomics is the study and identification of genomic markers that are associated with variability in cardiovascular drug response, cardiovascular drug-related outcomes, or cardiovascular drug-related adverse events. This overview presents an introduction and historical background to cardiovascular pharmacogenomics, and a protocol for designing a cardiovascular pharmacogenomics study. Important considerations are also included for constructing a cardiovascular pharmacogenomics phenotype, designing the replication or validation strategy, common statistical approaches, and how to put the results in context with the cardiovascular drug or cardiovascular disease under investigation. © 2021 Wiley Periodicals LLC. Basic Protocol: Designing a cardiovascular pharmacogenomics study.
Collapse
Affiliation(s)
- Caitrin W McDonough
- Department of Pharmacotherapy and Translational Research, College of Pharmacy, University of Florida, Gainesville, Florida
| |
Collapse
|
17
|
Fredette NC, Malik E, Mukhtar ML, Prossnitz ER, Terada N. A hypertension patient-derived iPSC model demonstrates a role for G protein-coupled estrogen receptor in hypertension risk and development. Am J Physiol Cell Physiol 2020; 319:C825-C838. [PMID: 32783656 DOI: 10.1152/ajpcell.00350.2019] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Hypertension (HTN) is a polyfactorial disease that can manifest severe cardiovascular pathologies such as heart failure or stroke. Genome-wide association studies (GWAS) of HTN indicate that single-nucleotide polymorphisms (SNPs) contribute to increased risk for HTN and resistance to some HTN drug regimens (Hiltunen TP et al., J Am Heart Assoc 4: e001521, 2015; Le MT et al., PLoS One 8: e52062, 2013; McDonough CW et al., J Hypertens 31: 698-704, 2013; Vandell AG et al., Hypertension 60: 957-964, 2012). However, cellular mechanistic insights of such SNPs remain largely unknown. Using a bank of induced pluripotent stem cells (iPSCs) derived from patients with HTN and CRISPR/Cas9-mediated gene-editing approach, we investigated the effects of a female HTN risk-associated SNP (rs1154431) of the G protein-coupled estrogen receptor (GPER) (Bassuk SS, Manson JE., Clin Chem 60: 68-77, 2014) in vascular endothelial cells. Although GPER1 deletion reduced endothelial nitric oxide synthase (eNOS) activation in iPSC-derived endothelial cells (iECs), the polymorphism itself did not significantly affect eNOS and NO production in a comparison of isogenic hemizygous iECs expressing either normal (P16) or HTN-associated (L16) GPER. Interestingly, we demonstrate for the first time that GPER plays a role in regulation of adhesion molecule expression and monocyte adhesion to iECs. Moreover, the L16 iECs had higher expression of inflammation genes than P16 iECs, implying that the risk variant may affect carrier individuals through increased inflammatory activity. This study further indicates that iPSCs are a useful platform for exploring mechanistic insights underlying hypertension GWAS endeavors.
Collapse
Affiliation(s)
- Natalie C Fredette
- Department of Pathology, Immunology and Experimental Medicine, University of Florida College of Medicine, Gainesville, Florida
| | - Eliyah Malik
- Department of Pathology, Immunology and Experimental Medicine, University of Florida College of Medicine, Gainesville, Florida
| | - Marah L Mukhtar
- Department of Mechanical & Aerospace Engineering, University of Florida Herbert Wertheim College of Engineering, Gainesville, Florida
| | - Eric R Prossnitz
- Department of Internal Medicine, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| | - Naohiro Terada
- Department of Pathology, Immunology and Experimental Medicine, University of Florida College of Medicine, Gainesville, Florida
| |
Collapse
|
18
|
Rysz J, Franczyk B, Rysz-Górzyńska M, Gluba-Brzózka A. Pharmacogenomics of Hypertension Treatment. Int J Mol Sci 2020; 21:ijms21134709. [PMID: 32630286 PMCID: PMC7369859 DOI: 10.3390/ijms21134709] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 06/21/2020] [Accepted: 06/26/2020] [Indexed: 12/12/2022] Open
Abstract
Hypertension is one of the strongest modifiable cardiovascular risk factors, affecting an increasing number of people worldwide. Apart from poor medication adherence, the low efficacy of some therapies could also be related to inter-individual genetic variability. Genetic studies of families revealed that heritability accounts for 30% to 50% of inter-individual variation in blood pressure (BP). Genetic factors not only affect blood pressure (BP) elevation but also contribute to inter-individual variability in response to antihypertensive treatment. This article reviews the recent pharmacogenomics literature concerning the key classes of antihypertensive drugs currently in use (i.e., diuretics, β-blockers, ACE inhibitors, ARB, and CCB). Due to the numerous studies on this topic and the sometimes-contradictory results within them, the presented data are limited to several selected SNPs that alter drug response. Genetic polymorphisms can influence drug responses through genes engaged in the pathogenesis of hypertension that are able to modify the effects of drugs, modifications in drug–gene mechanistic interactions, polymorphisms within drug-metabolizing enzymes, genes related to drug transporters, and genes participating in complex cascades and metabolic reactions. The results of numerous studies confirm that genotype-based antihypertension therapies are the most effective and may help to avoid the occurrence of major adverse events, as well as decrease the costs of treatment. However, the genetic heritability of drug response phenotypes seems to remain hidden in multigenic and multifactorial complex traits. Therefore, further studies are required to analyze all associations and formulate final genome-based treatment recommendations.
Collapse
Affiliation(s)
- Jacek Rysz
- Department of Nephrology, Hypertension and Family Medicine, Medical University of Lodz, 90-549 Lodz, Poland; (J.R.); (B.F.)
| | - Beata Franczyk
- Department of Nephrology, Hypertension and Family Medicine, Medical University of Lodz, 90-549 Lodz, Poland; (J.R.); (B.F.)
| | - Magdalena Rysz-Górzyńska
- Department of Ophthalmology and Visual Rehabilitation, Medical University of Lodz, 90-549 Lodz, Poland;
| | - Anna Gluba-Brzózka
- Department of Nephrology, Hypertension and Family Medicine, Medical University of Lodz, 90-549 Lodz, Poland; (J.R.); (B.F.)
- Correspondence:
| |
Collapse
|
19
|
de las Fuentes L, Sung YJ, Sitlani CM, Avery CL, Bartz TM, Keyser CD, Evans DS, Li X, Musani SK, Ruiter R, Smith AV, Sun F, Trompet S, Xu H, Arnett DK, Bis JC, Broeckel U, Busch EL, Chen YDI, Correa A, Cummings SR, Floyd JS, Ford I, Guo X, Harris TB, Ikram MA, Lange L, Launer LJ, Reiner AP, Schwander K, Smith NL, Sotoodehnia N, Stewart JD, Stott DJ, Stürmer T, Taylor KD, Uitterlinden A, Vasan RS, Wiggins KL, Cupples LA, Gudnason V, Heckbert SR, Jukema JW, Liu Y, Psaty BM, Rao DC, Rotter JI, Stricker B, Wilson JG, Whitsel EA. Genome-wide meta-analysis of variant-by-diuretic interactions as modulators of lipid traits in persons of European and African ancestry. THE PHARMACOGENOMICS JOURNAL 2020; 20:482-493. [PMID: 31806883 PMCID: PMC7260079 DOI: 10.1038/s41397-019-0132-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 11/13/2019] [Accepted: 11/20/2019] [Indexed: 01/11/2023]
Abstract
Hypertension (HTN) is a significant risk factor for cardiovascular morbidity and mortality. Metabolic abnormalities, including adverse cholesterol and triglycerides (TG) profiles, are frequent comorbid findings with HTN and contribute to cardiovascular disease. Diuretics, which are used to treat HTN and heart failure, have been associated with worsening of fasting lipid concentrations. Genome-wide meta-analyses with 39,710 European-ancestry (EA) individuals and 9925 African-ancestry (AA) individuals were performed to identify genetic variants that modify the effect of loop or thiazide diuretic use on blood lipid concentrations. Both longitudinal and cross sectional data were used to compute cohort-specific interaction results, which were then combined through meta-analysis in each ancestry. These ancestry-specific results were further combined through trans-ancestry meta-analysis. Analysis of EA data identified two genome-wide significant (p < 5 × 10-8) loci with single nucleotide variant (SNV)-loop diuretic interaction on TG concentrations (including COL11A1). Analysis of AA data identified one genome-wide significant locus adjacent to BMP2 with SNV-loop diuretic interaction on TG concentrations. Trans-ancestry analysis strengthened evidence of association for SNV-loop diuretic interaction at two loci (KIAA1217 and BAALC). There were few significant SNV-thiazide diuretic interaction associations on TG concentrations and for either diuretic on cholesterol concentrations. Several promising loci were identified that may implicate biologic pathways that contribute to adverse metabolic side effects from diuretic therapy.
Collapse
Affiliation(s)
- Lisa de las Fuentes
- Cardiovascular Division, Department of Medicine, Washington University, St. Louis, MO, USA.
| | - Y J Sung
- Division of Biostatistics, Washington University, St. Louis, MO, USA
| | - C M Sitlani
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
| | - C L Avery
- Gillings School of Global Public Health, Department of Epidemiology, University of North Carolina, Chapel Hill, NC, USA
| | - T M Bartz
- Cardiovascular Health Research Unit, Departments of Medicine and Biostatistics, University of Washington, Seattle, WA, USA
| | - C de Keyser
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - D S Evans
- Research Institute, California Pacific Medical Center, San Francisco, CA, USA
| | - X Li
- Institute for Translational Genomics and Population Sciences, Department of Pediatrics, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - S K Musani
- Jackson Heart Study, Department of Medicine, University of Mississippi Medical Center, Jackson, MS, USA
| | - R Ruiter
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - A V Smith
- Icelandic Heart Association, Kopavogur, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - F Sun
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - S Trompet
- Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands
- Department of Gerontology and Geriatrics, Leiden University Medical Center, Leiden, The Netherlands
| | - H Xu
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - D K Arnett
- Dean's Office, University of Kentucky College of Public Health, Lexington, KY, USA
| | - J C Bis
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
| | - U Broeckel
- Section of Genomic Pediatrics, Department of Pediatrics, Medicine and Physiology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - E L Busch
- Channing Division of Network Medicine, Department of Medicine, Harvard Medical School and Brigham and Women's Hospital, Boston, MA, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Y-D I Chen
- Institute for Translational Genomics and Population Sciences, Department of Pediatrics, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - A Correa
- Jackson Heart Study, Department of Medicine, University of Mississippi Medical Center, Jackson, MS, USA
| | - S R Cummings
- Research Institute, California Pacific Medical Center, San Francisco, CA, USA
| | - J S Floyd
- Cardiovascular Health Research Unit, Departments of Medicine and Epidemiology, University of Washington, Seattle, WA, USA
| | - I Ford
- Robertson Center for biostatistics, University of Glasgow, Glasgow, UK
| | - X Guo
- Institute for Translational Genomics and Population Sciences, Department of Pediatrics, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - T B Harris
- Laboratory of Epidemiology and Population Sciences, Intramural Research Program, National Institute on Aging, Bethesda, MD, USA
| | - M A Ikram
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - L Lange
- Department of Genetics, University of Colorado, Denver, Denver, CO, USA
| | - L J Launer
- Laboratory of Epidemiology and Population Sciences, Intramural Research Program, National Institute on Aging, Bethesda, MD, USA
| | - A P Reiner
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- School of Public Health, Department of Epidemiology, University of Washington, Seattle, WA, USA
| | - K Schwander
- Division of Biostatistics, Washington University, St. Louis, MO, USA
| | - N L Smith
- Cardiovascular Health Research Unit, Department of Epidemiology, University of Washington, Seattle, WA, USA
- Seattle Epidemiologic Research and Information Center (ERIC), VA Cooperative Studies Program, VA Puget Sound Health Care System, Seattle, WA, USA
| | - N Sotoodehnia
- Cardiovascular Health Research Unit, Departments of Medicine and Epidemiology, University of Washington, Seattle, WA, USA
- Cardiology, Department of Medicine, University of Washington, Seattle, WA, USA
| | - J D Stewart
- Gillings School of Global Public Health, Department of Epidemiology, University of North Carolina, Chapel Hill, NC, USA
- Carolina Population Center, University of North Carolina, Chapel Hill, NC, USA
| | - D J Stott
- Institute of cardiovascular and medical sciences, Faculty of Medicine, University of Glasgow, Glasgow, United Kingdom
| | - T Stürmer
- Gillings School of Global Public Health, Department of Epidemiology, University of North Carolina, Chapel Hill, NC, USA
- Center for Pharmacoepidemiology, University of North Carolina, Chapel Hill, NC, USA
| | - K D Taylor
- Institute for Translational Genomics and Population Sciences, Department of Pediatrics, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - A Uitterlinden
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - R S Vasan
- The Framingham Heart Study, Framingham, MA, USA
- Department of Medicine, Boston University School of Medicine, Boston, MA, USA
| | - K L Wiggins
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
| | - L A Cupples
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
- The Framingham Heart Study, Framingham, MA, USA
| | - V Gudnason
- Icelandic Heart Association, Kopavogur, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - S R Heckbert
- Cardiovascular Health Research Unit, Department of Epidemiology, University of Washington, Seattle, WA, USA
| | - J W Jukema
- Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands
- Interuniversity Cardiology Institute of the Netherlands, Utrecht, The Netherlands
| | - Y Liu
- Division of Public Health Sciences, Department of Epidemiology and Prevention, Wake Forest University, Winston-, Salem, NC, USA
| | - B M Psaty
- Cardiovascular Health Research Unit, Departments of Epidemiology, Medicine, and Health Services, University of Washington, Seattle, WA, USA
- Kaiser Permanente Washington Health Research Institute, Seattle, WA, USA
| | - D C Rao
- Division of Biostatistics, Washington University, St. Louis, MO, USA
| | - J I Rotter
- Institute for Translational Genomics and Population Sciences, Department of Pediatrics, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - B Stricker
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - J G Wilson
- Biophysics and Physiology, Department of Medicine, University of Mississippi Medical Center, Jackson, MS, USA
| | - E A Whitsel
- Gillings School of Global Public Health, Department of Epidemiology, University of North Carolina, Chapel Hill, NC, USA
- School of Medicine, Department of Medicine, University of North Carolina, Chapel Hill, NC, USA
| |
Collapse
|
20
|
Tu J, Zhang B, Fang G, Chang W, Zhao Y. Neddylation-mediated Nedd4-2 activation regulates ubiquitination modification of renal NBCe1. Exp Cell Res 2020; 390:111958. [DOI: 10.1016/j.yexcr.2020.111958] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 03/10/2020] [Accepted: 03/11/2020] [Indexed: 12/11/2022]
|
21
|
Abstract
Hypertension is still the number one global killer. No matter what causes are, lowering blood pressure can significantly reduce cardiovascular complications, cardiovascular death, and total death. Unfortunately, some hypertensive individuals simply do not know having hypertension. Some knew it but either not being treated or treated but blood pressure does not achieve goal. The reasons for inadequate control of blood pressure are many. One important reason is that we are not very familiar with antihypertensive agents and less attention has been paid to comorbidities, complications as well as the hypertension-modified target organ damage in patients with hypertension. The right antihypertensive drug was not given to the right hypertensive patients at right time. This reviewer studied comprehensively the literature, hopefully that the review will help improve antihypertensive drug selection and antihypertensive therapy.
Collapse
Affiliation(s)
- Rutai Hui
- Chinese Academy of Medical Sciences FUWAI Hospital Hypertension Division, 167 Beilishilu West City District, 100037, Beijing People's Republic of China, China.
| |
Collapse
|
22
|
Oliveira-Paula GH, Pereira SC, Tanus-Santos JE, Lacchini R. Pharmacogenomics And Hypertension: Current Insights. PHARMACOGENOMICS & PERSONALIZED MEDICINE 2019; 12:341-359. [PMID: 31819590 PMCID: PMC6878918 DOI: 10.2147/pgpm.s230201] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 09/07/2019] [Accepted: 11/05/2019] [Indexed: 11/23/2022]
Abstract
Hypertension is a multifactorial disease that affects approximately one billion subjects worldwide and is a major risk factor associated with cardiovascular events, including coronary heart disease and cerebrovascular accidents. Therefore, adequate blood pressure control is important to prevent these events, reducing premature mortality and disability. However, only one third of patients have the effective control of blood pressure, despite several classes of antihypertensive drugs available. These disappointing outcomes may be at least in part explained by interpatient variability in drug response due to genetic polymorphisms. To address the effects of genetic polymorphisms on blood pressure responses to the antihypertensive drug classes, studies have applied candidate genes and genome wide approaches. More recently, a third approach that considers gene-gene interactions has also been applied in hypertension pharmacogenomics. In this article, we carried out a comprehensive review of recent findings on the pharmacogenomics of antihypertensive drugs, including diuretics, β-blockers, angiotensin-converting enzyme inhibitors and angiotensin II receptor blockers, and calcium channel blockers. We also discuss the limitations and inconsistences that have been found in hypertension pharmacogenomics and the challenges to implement this valuable approach in clinical practice.
Collapse
Affiliation(s)
- Gustavo H Oliveira-Paula
- Department of Medicine, Division of Cardiology, Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, New York, NY, USA.,Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP, Brazil
| | - Sherliane C Pereira
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP, Brazil
| | - Jose E Tanus-Santos
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP, Brazil
| | - Riccardo Lacchini
- Department of Psychiatric Nursing and Human Sciences, Ribeirao Preto College of Nursing, University of Sao Paulo, Ribeirao Preto, SP, Brazil
| |
Collapse
|
23
|
Sung YJ, de las Fuentes L, Winkler TW, Chasman DI, Bentley AR, Kraja AT, Ntalla I, Warren HR, Guo X, Schwander K, Manning AK, Brown MR, Aschard H, Feitosa MF, Franceschini N, Lu Y, Cheng CY, Sim X, Vojinovic D, Marten J, Musani SK, Kilpeläinen TO, Richard MA, Aslibekyan S, Bartz TM, Dorajoo R, Li C, Liu Y, Rankinen T, Smith AV, Tajuddin SM, Tayo BO, Zhao W, Zhou Y, Matoba N, Sofer T, Alver M, Amini M, Boissel M, Chai JF, Chen X, Divers J, Gandin I, Gao C, Giulianini F, Goel A, Harris SE, Hartwig FP, He M, Horimoto ARVR, Hsu FC, Jackson AU, Kammerer CM, Kasturiratne A, Komulainen P, Kühnel B, Leander K, Lee WJ, Lin KH, Luan J, Lyytikäinen LP, McKenzie CA, Nelson CP, Noordam R, Scott RA, Sheu WHH, Stančáková A, Takeuchi F, van der Most PJ, Varga TV, Waken RJ, Wang H, Wang Y, Ware EB, Weiss S, Wen W, Yanek LR, Zhang W, Zhao JH, Afaq S, Alfred T, Amin N, Arking DE, Aung T, Barr RG, Bielak LF, Boerwinkle E, Bottinger EP, Braund PS, Brody JA, Broeckel U, Cade B, Campbell A, Canouil M, Chakravarti A, Cocca M, Collins FS, Connell JM, de Mutsert R, de Silva HJ, et alSung YJ, de las Fuentes L, Winkler TW, Chasman DI, Bentley AR, Kraja AT, Ntalla I, Warren HR, Guo X, Schwander K, Manning AK, Brown MR, Aschard H, Feitosa MF, Franceschini N, Lu Y, Cheng CY, Sim X, Vojinovic D, Marten J, Musani SK, Kilpeläinen TO, Richard MA, Aslibekyan S, Bartz TM, Dorajoo R, Li C, Liu Y, Rankinen T, Smith AV, Tajuddin SM, Tayo BO, Zhao W, Zhou Y, Matoba N, Sofer T, Alver M, Amini M, Boissel M, Chai JF, Chen X, Divers J, Gandin I, Gao C, Giulianini F, Goel A, Harris SE, Hartwig FP, He M, Horimoto ARVR, Hsu FC, Jackson AU, Kammerer CM, Kasturiratne A, Komulainen P, Kühnel B, Leander K, Lee WJ, Lin KH, Luan J, Lyytikäinen LP, McKenzie CA, Nelson CP, Noordam R, Scott RA, Sheu WHH, Stančáková A, Takeuchi F, van der Most PJ, Varga TV, Waken RJ, Wang H, Wang Y, Ware EB, Weiss S, Wen W, Yanek LR, Zhang W, Zhao JH, Afaq S, Alfred T, Amin N, Arking DE, Aung T, Barr RG, Bielak LF, Boerwinkle E, Bottinger EP, Braund PS, Brody JA, Broeckel U, Cade B, Campbell A, Canouil M, Chakravarti A, Cocca M, Collins FS, Connell JM, de Mutsert R, de Silva HJ, Dörr M, Duan Q, Eaton CB, Ehret G, Evangelou E, Faul JD, Forouhi NG, Franco OH, Friedlander Y, Gao H, Gigante B, Gu CC, Gupta P, Hagenaars SP, Harris TB, He J, Heikkinen S, Heng CK, Hofman A, Howard BV, Hunt SC, Irvin MR, Jia Y, Katsuya T, Kaufman J, Kerrison ND, Khor CC, Koh WP, Koistinen HA, Kooperberg CB, Krieger JE, Kubo M, Kutalik Z, Kuusisto J, Lakka TA, Langefeld CD, Langenberg C, Launer LJ, Lee JH, Lehne B, Levy D, Lewis CE, Li Y, Lim SH, Liu CT, Liu J, Liu J, Liu Y, Loh M, Lohman KK, Louie T, Mägi R, Matsuda K, Meitinger T, Metspalu A, Milani L, Momozawa Y, Mosley, Jr TH, Nalls MA, Nasri U, O'Connell JR, Ogunniyi A, Palmas WR, Palmer ND, Pankow JS, Pedersen NL, Peters A, Peyser PA, Polasek O, Porteous D, Raitakari OT, Renström F, Rice TK, Ridker PM, Robino A, Robinson JG, Rose LM, Rudan I, Sabanayagam C, Salako BL, Sandow K, Schmidt CO, Schreiner PJ, Scott WR, Sever P, Sims M, Sitlani CM, Smith BH, Smith JA, Snieder H, Starr JM, Strauch K, Tang H, Taylor KD, Teo YY, Tham YC, Uitterlinden AG, Waldenberger M, Wang L, Wang YX, Wei WB, Wilson G, Wojczynski MK, Xiang YB, Yao J, Yuan JM, Zonderman AB, Becker DM, Boehnke M, Bowden DW, Chambers JC, Chen YDI, Weir DR, de Faire U, Deary IJ, Esko T, Farrall M, Forrester T, Freedman BI, Froguel P, Gasparini P, Gieger C, Horta BL, Hung YJ, Jonas JB, Kato N, Kooner JS, Laakso M, Lehtimäki T, Liang KW, Magnusson PKE, Oldehinkel AJ, Pereira AC, Perls T, Rauramaa R, Redline S, Rettig R, Samani NJ, Scott J, Shu XO, van der Harst P, Wagenknecht LE, Wareham NJ, Watkins H, Wickremasinghe AR, Wu T, Kamatani Y, Laurie CC, Bouchard C, Cooper RS, Evans MK, Gudnason V, Hixson J, Kardia SLR, Kritchevsky SB, Psaty BM, van Dam RM, Arnett DK, Mook-Kanamori DO, Fornage M, Fox ER, Hayward C, van Duijn CM, Tai ES, Wong TY, Loos RJF, Reiner AP, Rotimi CN, Bierut LJ, Zhu X, Cupples LA, Province MA, Rotter JI, Franks PW, Rice K, Elliott P, Caulfield MJ, Gauderman WJ, Munroe PB, Rao DC, Morrison AC. A multi-ancestry genome-wide study incorporating gene-smoking interactions identifies multiple new loci for pulse pressure and mean arterial pressure. Hum Mol Genet 2019; 28:2615-2633. [PMID: 31127295 PMCID: PMC6644157 DOI: 10.1093/hmg/ddz070] [Show More Authors] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Revised: 03/25/2019] [Accepted: 03/26/2019] [Indexed: 12/24/2022] Open
Abstract
Elevated blood pressure (BP), a leading cause of global morbidity and mortality, is influenced by both genetic and lifestyle factors. Cigarette smoking is one such lifestyle factor. Across five ancestries, we performed a genome-wide gene-smoking interaction study of mean arterial pressure (MAP) and pulse pressure (PP) in 129 913 individuals in stage 1 and follow-up analysis in 480 178 additional individuals in stage 2. We report here 136 loci significantly associated with MAP and/or PP. Of these, 61 were previously published through main-effect analysis of BP traits, 37 were recently reported by us for systolic BP and/or diastolic BP through gene-smoking interaction analysis and 38 were newly identified (P < 5 × 10-8, false discovery rate < 0.05). We also identified nine new signals near known loci. Of the 136 loci, 8 showed significant interaction with smoking status. They include CSMD1 previously reported for insulin resistance and BP in the spontaneously hypertensive rats. Many of the 38 new loci show biologic plausibility for a role in BP regulation. SLC26A7 encodes a chloride/bicarbonate exchanger expressed in the renal outer medullary collecting duct. AVPR1A is widely expressed, including in vascular smooth muscle cells, kidney, myocardium and brain. FHAD1 is a long non-coding RNA overexpressed in heart failure. TMEM51 was associated with contractile function in cardiomyocytes. CASP9 plays a central role in cardiomyocyte apoptosis. Identified only in African ancestry were 30 novel loci. Our findings highlight the value of multi-ancestry investigations, particularly in studies of interaction with lifestyle factors, where genomic and lifestyle differences may contribute to novel findings.
Collapse
Affiliation(s)
- Yun Ju Sung
- Division of Biostatistics, Washington University School of Medicine, St. Louis, MO, USA
| | - Lisa de las Fuentes
- Division of Biostatistics, Washington University School of Medicine, St. Louis, MO, USA
- Cardiovascular Division, Department of Medicine, Washington University, St. Louis, MO, USA
| | - Thomas W Winkler
- Department of Genetic Epidemiology, University of Regensburg, Regensburg, Germany
| | - Daniel I Chasman
- Preventive Medicine, Brigham and Women’s Hospital, Boston, MA, USA
| | - Amy R Bentley
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Aldi T Kraja
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA
| | - Ioanna Ntalla
- Clinical Pharmacology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Helen R Warren
- Clinical Pharmacology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
- NIHR Barts Cardiovascular Biomedical Research Centre, Queen Mary University of London, London, London, UK
| | - Xiuqing Guo
- Division of Genomic Outcomes, Department of Pediatrics, The Institute for Translational Genomics and Population Sciences, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Karen Schwander
- Division of Biostatistics, Washington University School of Medicine, St. Louis, MO, USA
| | - Alisa K Manning
- Center for Human Genetics Research, Massachusetts General Hospital, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Michael R Brown
- Human Genetics Center, Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Hugues Aschard
- Department of Epidemiology, Harvard School of Public Health, Boston, MA, USA
- Centre de Bioinformatique, Biostatistique et Biologie Intégrative (C3BI), Institut Pasteur, Paris, France
| | - Mary F Feitosa
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA
| | - Nora Franceschini
- Epidemiology, University of North Carolina Gillings School of Global Public Health, Chapel Hill, NC, USA
| | - Yingchang Lu
- Icahn School of Medicine at Mount Sinai, The Charles Bronfman Institute for Personalized Medicine, New York, NY, USA
| | - Ching-Yu Cheng
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore, Singapore
- Ophthalmology & Visual Sciences Academic Clinical Program (Eye ACP), Duke-NUS Medical School, Singapore, Singapore
- Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Xueling Sim
- Saw Swee Hock School of Public Health, National University Health System and National University of Singapore, Singapore, Singapore
| | - Dina Vojinovic
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Jonathan Marten
- Medical Research Council Human Genetics Unit, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK
| | - Solomon K Musani
- Jackson Heart Study, Department of Medicine, University of Mississippi Medical Center, Jackson, MS, USA
| | - Tuomas O Kilpeläinen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Environmental Medicine and Public Health, The Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Melissa A Richard
- Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Stella Aslibekyan
- Department of Epidemiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Traci M Bartz
- Cardiovascular Health Research Unit, Biostatistics and Medicine, University of Washington, Seattle, WA, USA
| | - Rajkumar Dorajoo
- Genome Institute of Singapore, Agency for Science Technology and Research, Singapore, Singapore
| | - Changwei Li
- Epidemiology and Biostatistics, University of Georgia at Athens College of Public Health, Athens, GA, USA
| | - Yongmei Liu
- Public Health Sciences, Epidemiology and Prevention, Wake Forest University Health Sciences, Winston-Salem, NC, USA
| | - Tuomo Rankinen
- Human Genomics Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA, USA
| | - Albert Vernon Smith
- Icelandic Heart Association, Kopavogur, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - Salman M Tajuddin
- Health Disparities Research Section, Laboratory of Epidemiology and Population Sciences, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Bamidele O Tayo
- Department of Public Health Sciences, Loyola University Chicago, Maywood, IL, USA
| | - Wei Zhao
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Yanhua Zhou
- Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Nana Matoba
- Laboratory for Statistical Analysis, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Tamar Sofer
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Division of Sleep and Circadian Disorders, Brigham and Women’s Hospital, Boston, MA, USA
| | - Maris Alver
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Marzyeh Amini
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen RB, The Netherlands
| | - Mathilde Boissel
- CNRS UMR 8199, European Genomic Institute for Diabetes (EGID), Institut Pasteur de Lille, University of Lille, Lille, France
| | - Jin Fang Chai
- Saw Swee Hock School of Public Health, National University Health System and National University of Singapore, Singapore, Singapore
| | - Xu Chen
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Stockholm, Sweden
| | - Jasmin Divers
- Biostatistical Sciences, Public Health Sciences, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Ilaria Gandin
- Department of Medical Sciences, University of Trieste, Trieste, Italy
| | - Chuan Gao
- Molecular Genetics and Genomics Program, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | | | - Anuj Goel
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, Oxfordshire, UK
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, Oxfordshire, UK
| | - Sarah E Harris
- Centre for Cognitive Ageing and Cognitive Epidemiology, The University of Edinburgh, Edinburgh, UK
- Medical Genetics Section, University of Edinburgh Centre for Genomic and Experimental Medicine and MRC Institute of Genetics and Molecular Medicine, The University of Edinburgh, Edinburgh, UK
| | - Fernando P Hartwig
- Postgraduate Programme in Epidemiology, Federal University of Pelotas, Pelotas, RS, Brazil
- Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, UK
| | - Meian He
- Lab Genetics and Molecular Cardiology, Cardiology, Heart Institute, University of Sao Paulo, Sao Paulo, CA, USA
| | - Andrea R V R Horimoto
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, MI, USA
| | - Fang-Chi Hsu
- Biostatistical Sciences, Public Health Sciences, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Anne U Jackson
- Department of Human Genetics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Candace M Kammerer
- Department of Public Health, Faculty of Medicine, University of Kelaniya, Ragama, Sri Lanka
| | - Anuradhani Kasturiratne
- Foundation for Research in Health Exercise and Nutrition, Kuopio Research Institute of Exercise Medicine, Kuopio, Finland
| | - Pirjo Komulainen
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Brigitte Kühnel
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Karin Leander
- Medical Research, Taichung Veterans General Hospital, Department of Social Work, Tunghai University, Taichung, Taiwan
| | - Wen-Jane Lee
- Ophthalmology, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Keng-Hung Lin
- MRC Epidemiology Unit, University of Cambridge, Cambridge, UK
| | - Jian’an Luan
- Department of Clinical Chemistry, Fimlab Laboratories, Tampere, Finland
| | - Leo-Pekka Lyytikäinen
- Department of Clinical Chemistry, Finnish Cardiovascular Research Center—Tampere, Faculty of Medicine and Life Sciences, University of Tampere, Tampere, Finland
- Tropical Metabolism Research Unit, Tropical Medicine Research Institute, University of the West Indies, Mona, Jamaica
| | - Colin A McKenzie
- School of Public Health, Department of Occupational and Environmental Health and State Key Laboratory of Environmental Health for Incubating, Tongi Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Christopher P Nelson
- Department of Cardiovascular Sciences, University of Leicester, Leicester, UK
- NIHR Leicester Biomedical Research Centre, Glenfield Hospital, Leicester, UK
| | - Raymond Noordam
- Internal Medicine, Gerontology and Geriatrics, Leiden University Medical Center, Leiden, The Netherlands
| | - Robert A Scott
- Department of Clinical Chemistry, Fimlab Laboratories, Tampere, Finland
| | - Wayne H H Sheu
- Endocrinology and Metabolism, Internal Medicine, Taichung Veterans General Hospital, Taichung, Taiwan
- School of Medicine, National Yang-ming University, Taipei, Taiwan
- School of Medicine, National Defense Medical Center, Taipei, Taiwan
- Institute of Medical Technology, National Chung-Hsing University, Taichung, Taiwan
| | - Alena Stančáková
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland, Kuopio, Finland
| | - Fumihiko Takeuchi
- Department of Gene Diagnostics and Therapeutics, Research Institute, National Center for Global Health and Medicine, Tokyo, Japan
| | - Peter J van der Most
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen RB, The Netherlands
| | - Tibor V Varga
- Department of Clinical Sciences, Genetic and Molecular Epidemiology Unit, Lund University Diabetes Centre, Skåne University Hospital, Malmö, Sweden
| | - Robert J Waken
- Division of Biostatistics, Washington University School of Medicine, St. Louis, MO, USA
| | - Heming Wang
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Division of Sleep and Circadian Disorders, Brigham and Women’s Hospital, Boston, MA, USA
| | - Yajuan Wang
- Department of Epidemiology and Biostatistics, Case Western Reserve University, Cleveland, OH, USA
| | - Erin B Ware
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, USA
- Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, MI, USA
| | - Stefan Weiss
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine Ernst Moritz Arndt University Greifswald, Greifswald, Germany
- DZHK (German Centre for Cardiovascular Health), Partner Site Greifswald, Greifswald, Germany
| | - Wanqing Wen
- Division of Epidemiology, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Lisa R Yanek
- General Internal Medicine, GeneSTAR Research Program, Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Weihua Zhang
- MRC-PHE Centre for Environment and Health, Department of Epidemiology and Biostatistics, Imperial College London, London, UK
- Department of Cardiology, Ealing Hospital, Middlesex, UK
| | - Jing Hua Zhao
- Department of Clinical Chemistry, Fimlab Laboratories, Tampere, Finland
| | - Saima Afaq
- MRC-PHE Centre for Environment and Health, Department of Epidemiology and Biostatistics, Imperial College London, London, UK
| | - Tamuno Alfred
- Icahn School of Medicine at Mount Sinai, The Charles Bronfman Institute for Personalized Medicine, New York, NY, USA
| | - Najaf Amin
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Dan E Arking
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Tin Aung
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore, Singapore
- Ophthalmology & Visual Sciences Academic Clinical Program (Eye ACP), Duke-NUS Medical School, Singapore, Singapore
- Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - R Graham Barr
- Departments of Medicine and Epidemiology, Columbia University Medical Center, New York, NY, USA
| | - Lawrence F Bielak
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Eric Boerwinkle
- Human Genetics Center, Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, USA
| | - Erwin P Bottinger
- Icahn School of Medicine at Mount Sinai, The Charles Bronfman Institute for Personalized Medicine, New York, NY, USA
| | - Peter S Braund
- Department of Cardiovascular Sciences, University of Leicester, Leicester, UK
- NIHR Leicester Biomedical Research Centre, Glenfield Hospital, Leicester, UK
| | - Jennifer A Brody
- Cardiovascular Health Research Unit, Medicine, University of Washington, Seattle, WA, USA
| | - Ulrich Broeckel
- Section of Genomic Pediatrics, Department of Pediatrics, Medicine and Physiology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Brian Cade
- Division of Sleep and Circadian Disorders, Brigham and Women’s Hospital, Boston, MA, USA
| | - Archie Campbell
- Centre for Genomic & Experimental Medicine, Institute of Genetics & Molecular Medicine, University of Edinburgh, Edinburgh, UK
| | - Mickaël Canouil
- CNRS UMR 8199, European Genomic Institute for Diabetes (EGID), Institut Pasteur de Lille, University of Lille, Lille, France
| | - Aravinda Chakravarti
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | - Francis S Collins
- Medical Genomics and Metabolic Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - John M Connell
- Ninewells Hospital & Medical School, University of Dundee, Dundee, Scotland, UK
| | - Renée de Mutsert
- Clinical Epidemiology, Leiden University Medical Center, Leiden, The Netherlands
| | - H Janaka de Silva
- Department of Medicine, Faculty of Medicine, University of Kelaniya, Ragama, Sri Lanka
| | - Marcus Dörr
- DZHK (German Centre for Cardiovascular Health), Partner Site Greifswald, Greifswald, Germany
- Department of Internal Medicine B, University Medicine Greifswald, Greifswald, Germany
| | - Qing Duan
- Department of Genetics, University of North Carolina, Chapel Hill, USA
| | - Charles B Eaton
- Department of Family Medicine and Epidemiology, Alpert Medical School of Brown University, Providence, RI, USA
| | - Georg Ehret
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Cardiology, Department of Specialties of Medicine, Geneva University Hospital, Geneva, Switzerland
| | - Evangelos Evangelou
- MRC-PHE Centre for Environment and Health, Department of Epidemiology and Biostatistics, Imperial College London, London, UK
- Department of Hygiene and Epidemiology, University of Ioannina Medical School, Ioannina, Greece
| | - Jessica D Faul
- Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, MI, USA
| | - Nita G Forouhi
- Department of Clinical Chemistry, Fimlab Laboratories, Tampere, Finland
| | - Oscar H Franco
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Yechiel Friedlander
- Braun School of Public Health, Hebrew University-Hadassah Medical Center, Jerusalem, Israel
| | - He Gao
- MRC-PHE Centre for Environment and Health, Department of Epidemiology and Biostatistics, Imperial College London, London, UK
| | - Bruna Gigante
- Medical Research, Taichung Veterans General Hospital, Department of Social Work, Tunghai University, Taichung, Taiwan
| | - C Charles Gu
- Division of Biostatistics, Washington University School of Medicine, St. Louis, MO, USA
| | - Preeti Gupta
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore, Singapore
| | - Saskia P Hagenaars
- Centre for Cognitive Ageing and Cognitive Epidemiology, The University of Edinburgh, Edinburgh, UK
- Psychology, The University of Edinburgh, Edinburgh, UK
| | - Tamara B Harris
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| | - Jiang He
- Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA, USA
- Medicine, Tulane University School of Medicine, New Orleans, LA, USA
| | - Sami Heikkinen
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland, Kuopio, Finland
- Institute of Biomedicine, School of Medicine, University of Eastern Finland, Kuopio Campus, Finland
| | - Chew-Kiat Heng
- Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Khoo Teck Puat—National University Children’s Medical Institute, National University Health System, Singapore, Singapore
| | - Albert Hofman
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Barbara V Howard
- MedStar Health Research Institute, Hyattsville, MD, USA
- Center for Clinical and Translational Sciences and Department of Medicine, Georgetown–Howard Universities, Washington, DC, USA
| | - Steven C Hunt
- Division of Epidemiology, Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, UT, USA
- Department of Genetic Medicine, Weill Cornell Medicine, Doha, Qatar
| | - Marguerite R Irvin
- Department of Biostatistics, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Yucheng Jia
- Division of Genomic Outcomes, Department of Pediatrics, The Institute for Translational Genomics and Population Sciences, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Tomohiro Katsuya
- Department of Clinical Gene Therapy, Osaka University Graduate School of Medicine, Suita, Japan
- Department of Geriatric Medicine and Nephrology, Osaka University Graduate School of Medicine, Suita, Japan
| | - Joel Kaufman
- Epidemiology, Occupational and Environmental Medicine Program, University of Washington, Seattle, WA, USA
| | - Nicola D Kerrison
- Department of Clinical Chemistry, Fimlab Laboratories, Tampere, Finland
| | - Chiea Chuen Khor
- Genome Institute of Singapore, Agency for Science Technology and Research, Singapore, Singapore
- Department of Biochemistry, National University of Singapore, Singapore, Singapore
| | - Woon-Puay Koh
- Saw Swee Hock School of Public Health, National University Health System and National University of Singapore, Singapore, Singapore
- Health Services and Systems Research, Duke–NUS Medical School, Singapore, Singapore
| | - Heikki A Koistinen
- Department of Public Health Solutions, National Institute for Health and Welfare, Helsinki, Finland
- Department of Medicine and Abdominal Center: Endocrinology, University of Helsinki and Helsinki University Central Hospital, Helsinki, Finland
- Minerva Foundation Institute for Medical Research, Biomedicum 2U, Helsinki Finland
| | - Charles B Kooperberg
- Fred Hutchinson Cancer Research Center, University of Washington School of Public Health, Seattle, WA, USA
| | - Jose E Krieger
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, MI, USA
| | - Michiaki Kubo
- RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Zoltan Kutalik
- Institute of Social Preventive Medicine, Lausanne University Hospital, Lausanne, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Johanna Kuusisto
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland, Kuopio, Finland
| | - Timo A Lakka
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Institute of Biomedicine, School of Medicine, University of Eastern Finland, Kuopio Campus, Finland
- Department of Clinical Physiology and Nuclear Medicine, Kuopio University Hospital, Kuopio, Finland
| | - Carl D Langefeld
- Biostatistical Sciences, Public Health Sciences, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | | | - Lenore J Launer
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| | - Joseph H Lee
- Sergievsky Center, College of Physicians and Surgeons, Columbia University Mailman School of Public Health, New York, NY, USA
| | - Benjamin Lehne
- MRC-PHE Centre for Environment and Health, Department of Epidemiology and Biostatistics, Imperial College London, London, UK
| | - Daniel Levy
- NHLBI Framingham Heart Study, Framingham, MA, USA
- The Population Sciences Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Cora E Lewis
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Yize Li
- Division of Biostatistics, Washington University School of Medicine, St. Louis, MO, USA
| | | | - Sing Hui Lim
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore, Singapore
| | - Ching-Ti Liu
- Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Jianjun Liu
- Saw Swee Hock School of Public Health, National University Health System and National University of Singapore, Singapore, Singapore
- Genome Institute of Singapore, Agency for Science Technology and Research, Singapore, Singapore
| | - Jingmin Liu
- WHI CCC, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Yeheng Liu
- Division of Genomic Outcomes, Department of Pediatrics, The Institute for Translational Genomics and Population Sciences, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Marie Loh
- MRC-PHE Centre for Environment and Health, Department of Epidemiology and Biostatistics, Imperial College London, London, UK
- Translational Laboratory in Genetic Medicine, Agency for Science, Technology and Research, Singapore
| | - Kurt K Lohman
- Biostatistical Sciences, Public Health Sciences, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Tin Louie
- Department of Biostatistics, University of Washington, Seattle, WA, USA
| | - Reedik Mägi
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Koichi Matsuda
- Laboratory for Clinical Genome Sequencing, Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Minato-ku, Japan
| | - Thomas Meitinger
- Institute of Human Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Institute of Human Genetics, Technische Universität München, Munich, Germany
| | - Andres Metspalu
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Lili Milani
- Cardiovascular Division, Department of Medicine, Washington University, St. Louis, MO, USA
| | - Yukihide Momozawa
- Laboratory for Genotyping Development, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | | | - Mike A Nalls
- Laboratory of Neurogenetics, National Institute on Aging, Bethesda, MD, USA
| | - Ubaydah Nasri
- Division of Genomic Outcomes, Department of Pediatrics, The Institute for Translational Genomics and Population Sciences, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Jeff R O'Connell
- Division of Endocrinology, Diabetes, and Nutrition, University of Maryland School of Medicine, Baltimore, MD, USA
- Program for Personalized and Genomic Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | | | | | | | - James S Pankow
- Division of Epidemiology and Community Health, University of Minnesota School of Public Health, Minneapolis, MN, USA
| | - Nancy L Pedersen
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Stockholm, Sweden
| | - Annette Peters
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Neuherberg, Germany
| | - Patricia A Peyser
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Ozren Polasek
- Department of Public Health, Department of Medicine, University of Split, Split, Croatia
- Psychiatric Hospital ‘Sveti Ivan’, Zagreb, Croatia
- Gen-info Ltd, Zagreb, Croatia
| | - David Porteous
- Centre for Genomic & Experimental Medicine, Institute of Genetics & Molecular Medicine, University of Edinburgh, Edinburgh, UK
| | - Olli T Raitakari
- Department of Clinical Physiology and Nuclear Medicine, Turku University Hospital, Turku, Finland
- Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, Turku, Finland
| | - Frida Renström
- Department of Clinical Sciences, Genetic and Molecular Epidemiology Unit, Lund University Diabetes Centre, Skåne University Hospital, Malmö, Sweden
- Department of Biobank Research, Umeå University, Umeå, Västerbotten, Sweden
| | - Treva K Rice
- Division of Biostatistics, Washington University School of Medicine, St. Louis, MO, USA
| | - Paul M Ridker
- Preventive Medicine, Brigham and Women’s Hospital, Boston, MA, USA
| | - Antonietta Robino
- Institute for Maternal and Child Health—IRCCS ‘Burlo Garofolo’, Trieste, Italy
| | - Jennifer G Robinson
- Department of Epidemiology and Medicine, University of Iowa, Iowa City, IA, USA
| | - Lynda M Rose
- Preventive Medicine, Brigham and Women’s Hospital, Boston, MA, USA
| | - Igor Rudan
- Centre for Global Health Research, Usher Institute of Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, UK
| | - Charumathi Sabanayagam
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore, Singapore
- Ophthalmology & Visual Sciences Academic Clinical Program (Eye ACP), Duke-NUS Medical School, Singapore, Singapore
| | | | - Kevin Sandow
- Division of Genomic Outcomes, Department of Pediatrics, The Institute for Translational Genomics and Population Sciences, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Carsten O Schmidt
- DZHK (German Centre for Cardiovascular Health), Partner Site Greifswald, Greifswald, Germany
- Institute for Community Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Pamela J Schreiner
- Division of Epidemiology and Community Health, University of Minnesota School of Public Health, Minneapolis, MN, USA
| | - William R Scott
- MRC-PHE Centre for Environment and Health, Department of Epidemiology and Biostatistics, Imperial College London, London, UK
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Peter Sever
- International Centre for Circulatory Health, Imperial College London, London, UK
| | - Mario Sims
- Jackson Heart Study, Department of Medicine, University of Mississippi Medical Center, Jackson, MS, USA
| | - Colleen M Sitlani
- Cardiovascular Health Research Unit, Medicine, University of Washington, Seattle, WA, USA
| | - Blair H Smith
- Division of Population Health Sciences, Ninewells Hospital and Medical School, University of Dundee, Dundee, UK
| | - Jennifer A Smith
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, USA
- Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, MI, USA
| | - Harold Snieder
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen RB, The Netherlands
| | - John M Starr
- Centre for Cognitive Ageing and Cognitive Epidemiology, The University of Edinburgh, Edinburgh, UK
- Alzheimer Scotland Dementia Research Centre, The University of Edinburgh, Edinburgh, UK
| | - Konstantin Strauch
- Institute of Genetic Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Genetic Epidemiology, IBE, Faculty of Medicine, LMU, Munich, Germany
| | - Hua Tang
- Department of Genetics, Stanford University, Stanford, CA, USA
| | - Kent D Taylor
- Division of Genomic Outcomes, Department of Pediatrics, The Institute for Translational Genomics and Population Sciences, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Yik Ying Teo
- Saw Swee Hock School of Public Health, National University Health System and National University of Singapore, Singapore, Singapore
- Genome Institute of Singapore, Agency for Science Technology and Research, Singapore, Singapore
- Life Sciences Institute, National University of Singapore, Singapore, Singapore
- NUS Graduate School for Integrative Science and Engineering, National University of Singapore, Singapore, Singapore
- Department of Statistics and Applied Probability, National University of Singapore, Singapore, Singapore
| | - Yih Chung Tham
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore, Singapore
| | - André G Uitterlinden
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Melanie Waldenberger
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- MRC Epidemiology Unit, University of Cambridge, Cambridge, UK
| | - Lihua Wang
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA
| | - Ya Xing Wang
- Beijing Institute of Ophthalmology, Beijing Tongren Eye Center, Beijing Ophthalmology and Visual Science Key Lab, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Wen Bin Wei
- Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Gregory Wilson
- Jackson Heart Study, School of Public Health, Jackson State University, Jackson, MS, USA
| | - Mary K Wojczynski
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA
| | - Yong-Bing Xiang
- State Key Laboratory of Oncogene and Related Genes & Department of Epidemiology, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, P.R. China
| | - Jie Yao
- Division of Genomic Outcomes, Department of Pediatrics, The Institute for Translational Genomics and Population Sciences, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Jian-Min Yuan
- Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
- Division of Cancer Control and Population Sciences, UPMC Hillman Cancer, University of Pittsburgh, Pittsburgh, PA, USA
| | - Alan B Zonderman
- Behavioral Epidemiology Section, Laboratory of Epidemiology and Population Sciences, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Diane M Becker
- General Internal Medicine, GeneSTAR Research Program, Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Michael Boehnke
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, MI, USA
| | - Donald W Bowden
- Biochemistry, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - John C Chambers
- MRC-PHE Centre for Environment and Health, Department of Epidemiology and Biostatistics, Imperial College London, London, UK
- Department of Cardiology, Ealing Hospital, Middlesex, UK
| | - Yii-Der Ida Chen
- Division of Genomic Outcomes, Department of Pediatrics, The Institute for Translational Genomics and Population Sciences, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - David R Weir
- Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, MI, USA
| | - Ulf de Faire
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Ian J Deary
- Centre for Cognitive Ageing and Cognitive Epidemiology, The University of Edinburgh, Edinburgh, UK
- Psychology, The University of Edinburgh, Edinburgh, UK
| | - Tõnu Esko
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu, Estonia
- Broad Institute of the Massachusetts Institute of Technology and Harvard University, Boston, MA, USA
| | - Martin Farrall
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, Oxfordshire, UK
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, Oxfordshire, UK
| | - Terrence Forrester
- Tropical Metabolism Research Unit, Tropical Medicine Research Institute, University of the West Indies, Mona, Jamaica
| | - Barry I Freedman
- Nephrology, Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Philippe Froguel
- CNRS UMR 8199, European Genomic Institute for Diabetes (EGID), Institut Pasteur de Lille, University of Lille, Lille, France
- Department of Genomics of Common Disease, Imperial College London, London, UK
| | - Paolo Gasparini
- Department of Medical Sciences, University of Trieste, Trieste, Italy
- Department of Genetic Medicine, Weill Cornell Medicine, Doha, Qatar
| | - Christian Gieger
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany
| | - Bernardo Lessa Horta
- Postgraduate Programme in Epidemiology, Federal University of Pelotas, Pelotas, RS, Brazil
| | - Yi-Jen Hung
- Endocrinology and Metabolism, Tri-Service General Hospital, National Defense Medical Center, Taipei City, Taipei, Taiwan
| | - Jost Bruno Jonas
- Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing, China
- Department of Ophthalmology, Medical Faculty Mannheim, University Heidelberg, Mannheim, Germany
| | - Norihiro Kato
- Department of Gene Diagnostics and Therapeutics, Research Institute, National Center for Global Health and Medicine, Tokyo, Japan
| | - Jaspal S Kooner
- Department of Cardiology, Ealing Hospital, Middlesex, UK
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Markku Laakso
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland, Kuopio, Finland
| | - Terho Lehtimäki
- Department of Clinical Chemistry, Fimlab Laboratories, Tampere, Finland
- Department of Clinical Chemistry, Finnish Cardiovascular Research Center—Tampere, Faculty of Medicine and Life Sciences, University of Tampere, Tampere, Finland
| | - Kae-Woei Liang
- School of Medicine, National Yang-ming University, Taipei, Taiwan
- Cardiovascular Center, Taichung Veterans General Hospital, Taichung, Taiwan
- Department of Medicine, China Medical University, Taichung, Taiwan
| | - Patrik K E Magnusson
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Stockholm, Sweden
| | - Albertine J Oldehinkel
- Department of Psychiatry, University of Groningen, University Medical Center Groningen, Groningen RB, The Netherlands
| | - Alexandre C Pereira
- Lab Genetics and Molecular Cardiology, Cardiology, Heart Institute, University of Sao Paulo, Sao Paulo, CA, USA
- Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Thomas Perls
- Geriatrics Section, Boston University Medical Center, Boston, MA, USA
| | - Rainer Rauramaa
- Foundation for Research in Health Exercise and Nutrition, Kuopio Research Institute of Exercise Medicine, Kuopio, Finland
| | - Susan Redline
- Division of Sleep and Circadian Disorders, Brigham and Women’s Hospital, Boston, MA, USA
| | - Rainer Rettig
- DZHK (German Centre for Cardiovascular Health), Partner Site Greifswald, Greifswald, Germany
- Institute of Physiology, University of Medicine Greifswald, Greifswald, Germany
| | - Nilesh J Samani
- Department of Cardiovascular Sciences, University of Leicester, Leicester, UK
- NIHR Leicester Biomedical Research Centre, Glenfield Hospital, Leicester, UK
| | - James Scott
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Xiao-Ou Shu
- Division of Epidemiology, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Pim van der Harst
- Department of Cardiology, University of Groningen, University Medical Center Groningen, Groningen RB, The Netherlands
| | - Lynne E Wagenknecht
- Public Health Sciences, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | | | - Hugh Watkins
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, Oxfordshire, UK
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, Oxfordshire, UK
| | | | - Tangchun Wu
- School of Public Health, Department of Occupational and Environmental Health and State Key Laboratory of Environmental Health for Incubating, Tongi Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yoichiro Kamatani
- Laboratory for Statistical Analysis, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Cathy C Laurie
- Department of Biostatistics, University of Washington, Seattle, WA, USA
| | - Claude Bouchard
- Human Genomics Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA, USA
| | - Richard S Cooper
- Department of Public Health Sciences, Loyola University Chicago, Maywood, IL, USA
| | - Michele K Evans
- Health Disparities Research Section, Laboratory of Epidemiology and Population Sciences, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Vilmundur Gudnason
- Icelandic Heart Association, Kopavogur, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - James Hixson
- Human Genetics Center, Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Sharon L R Kardia
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Stephen B Kritchevsky
- Department of Internal Medicine, Section on Gerontology and Geriatric Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Bruce M Psaty
- Cardiovascular Health Research Unit, Epidemiology, Medicine and Health Services, University of Washington, Seattle, WA, USA
- Kaiser Permanente Washington Health Research Institute, Seattle, WA, USA
| | - Rob M van Dam
- Saw Swee Hock School of Public Health, National University Health System and National University of Singapore, Singapore, Singapore
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Donna K Arnett
- Dean’s Office, University of Kentucky College of Public Health, Lexington, KY, USA
| | - Dennis O Mook-Kanamori
- Clinical Epidemiology, Leiden University Medical Center, Leiden, The Netherlands
- Public Health and Primary Care, Leiden University Medical Center, Leiden, The Netherlands
| | - Myriam Fornage
- Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Ervin R Fox
- Cardiology, Medicine, University of Mississippi Medical Center, Jackson, MS, USA
| | - Caroline Hayward
- Medical Research Council Human Genetics Unit, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK
| | - Cornelia M van Duijn
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - E Shyong Tai
- Saw Swee Hock School of Public Health, National University Health System and National University of Singapore, Singapore, Singapore
- Health Services and Systems Research, Duke–NUS Medical School, Singapore, Singapore
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Tien Yin Wong
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore, Singapore
- Ophthalmology & Visual Sciences Academic Clinical Program (Eye ACP), Duke-NUS Medical School, Singapore, Singapore
- Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Ruth J F Loos
- Icahn School of Medicine at Mount Sinai, The Charles Bronfman Institute for Personalized Medicine, New York, NY, USA
- Icahn School of Medicine at Mount Sinai, The Mindich Child Health and Development Institute, New York, NY, USA
| | - Alex P Reiner
- Fred Hutchinson Cancer Research Center, University of Washington School of Public Health, Seattle, WA, USA
| | - Charles N Rotimi
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Laura J Bierut
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
| | - Xiaofeng Zhu
- Department of Epidemiology and Biostatistics, Case Western Reserve University, Cleveland, OH, USA
| | - L Adrienne Cupples
- Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Michael A Province
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA
| | - Jerome I Rotter
- Division of Genomic Outcomes, Department of Pediatrics, The Institute for Translational Genomics and Population Sciences, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Paul W Franks
- Department of Clinical Sciences, Genetic and Molecular Epidemiology Unit, Lund University Diabetes Centre, Skåne University Hospital, Malmö, Sweden
- Harvard T. H. Chan School of Public Health, Department of Nutrition, Harvard University, Boston, MA, USA
- Department of Public Health & Clinical Medicine, Umeå University, Umeå, Västerbotten, Sweden
| | - Kenneth Rice
- Department of Biostatistics, University of Washington, Seattle, WA, USA
| | - Paul Elliott
- MRC-PHE Centre for Environment and Health, Department of Epidemiology and Biostatistics, Imperial College London, London, UK
| | - Mark J Caulfield
- Clinical Pharmacology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
- NIHR Barts Cardiovascular Biomedical Research Centre, Queen Mary University of London, London, London, UK
| | - W James Gauderman
- Biostatistics, Preventive Medicine, University of Southern California, Los Angeles, CA, USA
| | - Patricia B Munroe
- Clinical Pharmacology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
- NIHR Barts Cardiovascular Biomedical Research Centre, Queen Mary University of London, London, London, UK
| | - Dabeeru C Rao
- Division of Biostatistics, Washington University School of Medicine, St. Louis, MO, USA
| | - Alanna C Morrison
- Human Genetics Center, Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
| |
Collapse
|
24
|
Wang X, Duan J, Fu W, Yin Z, Sheng J, Lei Z, Wang H. Decreased expression of NEDD4L contributes to NSCLC progression and metastasis. Biochem Biophys Res Commun 2019; 513:398-404. [PMID: 30967264 DOI: 10.1016/j.bbrc.2019.04.001] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Accepted: 04/01/2019] [Indexed: 01/22/2023]
Abstract
Recent evidence indicated that neural precursor cell expressed, developmentally down-regulated 4-like (NEDD4L) has a critical role in the regulation of cellular processes such as apoptosis, transport and metastasis, and is downregulated in several types of cancers. However, the role of NEDD4L in non-small cell lung cancer (NSCLC) has not been fully elucidated. In this study, we demonstrated that NEDD4L was downregulated in NSCLCs. This downregulation correlated with lymph node invasion, advanced stage and poor survival. In vitro experiments revealed that NEDD4L significantly suppressed cell proliferation, migration and invasion abilities. Further in vivo assay demonstrated that knocking down of NEDD4L enhanced the tumor metastasis of NSCLC cells. Moreover, we found that Polycomb group protein enhancer of zeste homologue 2 (EZH2) mediated H3K27 methylation was involved in the downregulation of NEDD4L. Knocking down of EZH2 restored the expression of NEDD4L. Further examined by luciferase reporter assay indicated the EZH2 regulated the transcription activity of NEDD4L. In clinical samples, EZH2 was inversely correlated with NEDD4L expression. In summary, NEDD4L acted as a tumor suppressor gene in NSCLC and targeting EZH2 could upregulate NEDD4L expression, which might serve as a novel approach for NSCLC.
Collapse
Affiliation(s)
- Xuming Wang
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Kunming Medical University, Kunming, 650032, China
| | - Jin Duan
- Department of Geriatric Thoracic Surgery, The First Affiliated Hospital of Kunming Medical University, Kunming, 650032, China
| | - Weiping Fu
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Kunming Medical University, Kunming, 650032, China
| | - Zhaowu Yin
- Department of Oncology, The People's Hospital of Tengchong County, Baoshan, 679100, China
| | - Jianing Sheng
- Department of Oncology, The People's Hospital of Tengchong County, Baoshan, 679100, China
| | - Zhuyun Lei
- Department of Oncology, The People's Hospital of Tengchong County, Baoshan, 679100, China.
| | - Han Wang
- Department of Thoracic Surgery, The First People's Hospital of Yunnan Province, Kunming, 650032, China.
| |
Collapse
|
25
|
Cunningham PN, Chapman AB. The future of pharmacogenetics in the treatment of hypertension. Pharmacogenomics 2019; 20:129-132. [PMID: 30808251 DOI: 10.2217/pgs-2018-0191] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
|
26
|
Disorders of renal NaCl transport and implications for blood pressure regulation. MED GENET-BERLIN 2019. [DOI: 10.1007/s11825-019-0232-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Abstract
Hypertension is one of the major risk factors for cardiovascular disease in industrialized societies. Substantial progress has been made in understanding its epidemiology, its pathophysiology, and its associated risks such as coronary artery disease, stroke, and heart failure. Because there is consensus that the abnormal retention of sodium by the kidney is a major important pathophysiological event in hypertension, this review focuses on mechanisms of renal NaCl transport and associated genetic disorders.
Collapse
|
27
|
Rodriguez-Iturbe B, Johnson RJ. Genetic Polymorphisms in Hypertension: Are We Missing the Immune Connection? Am J Hypertens 2019; 32:113-122. [PMID: 30418477 DOI: 10.1093/ajh/hpy168] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Accepted: 11/06/2018] [Indexed: 01/11/2023] Open
Affiliation(s)
- Bernardo Rodriguez-Iturbe
- Renal Hospital Universitario, Universidad del Zulia, Maracaibo, Zulia, Venezuela
- Instituto Venezolano de Investigaciones Científicas (IVIC)-Zulia, Maracaibo, Zulia, Venezuela
| | - Richard J Johnson
- Division of Renal Diseases and Hypertension, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| |
Collapse
|
28
|
Hoh BP, Abdul Rahman T, Yusoff K. Natural selection and local adaptation of blood pressure regulation and their perspectives on precision medicine in hypertension. Hereditas 2019; 156:1. [PMID: 30636949 PMCID: PMC6323824 DOI: 10.1186/s41065-019-0080-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2018] [Accepted: 01/01/2019] [Indexed: 01/09/2023] Open
Abstract
Prevalence of hypertension (HTN) varies substantially across different populations. HTN is not only common - affecting at least one third of the world's adult population - but is also the most important driver for cardiovascular diseases. Yet up to a third of hypertensive patients are resistant to therapy, contributed by secondary hypertension but more commonly the hitherto inability to precisely predict response to specific antihypertensive agents. Population and individual genomics information could be useful in guiding the selection and predicting the response to treatment - an approach known as precision medicine. However this cannot be achieved without the knowledge of genetic variations that influence blood pressure (BP). A number of evolutionary factors including population demographics and forces of natural selection may be involved. This article explores some ideas on how natural selection influences BP regulation in ethnically and geographically diverse populations that could lead to them being susceptible to HTN. We explore how such evolutionary factors could impact the implementation of precision medicine in HTN. Finally, in order to ensure the success of precision medicine in HTN, we call for more initiatives to understand the genetic architecture within and between diverse populations with ancestry from different parts of the world, and to precisely classify the intermediate phenotypes of HTN.
Collapse
Affiliation(s)
- Boon-Peng Hoh
- 1Faculty of Medicine and Health Sciences, UCSI University, Cheras, 56000 Kuala Lumpur, Malaysia.,2Chinese Academy of Sciences Key Laboratory of Computational Biology, Max Planck Independent Research Group on Population Genomics, CAS-MPG Partner Institute for Computational Biology, Shanghai Institutes for Biological Sciences, CAS, Shanghai, 200031 China
| | - Thuhairah Abdul Rahman
- 3Clinical Pathology Diagnostic Centre Research Laboratory, Faculty of Medicine, Universiti Teknologi MARA, Sungai Buloh Campus, 47000 Sungai Buloh, Selangor Malaysia
| | - Khalid Yusoff
- 1Faculty of Medicine and Health Sciences, UCSI University, Cheras, 56000 Kuala Lumpur, Malaysia
| |
Collapse
|
29
|
|
30
|
Dong OM. Excessive dietary sodium intake and elevated blood pressure: a review of current prevention and management strategies and the emerging role of pharmaconutrigenetics. BMJ Nutr Prev Health 2018; 1:7-16. [PMID: 33235949 PMCID: PMC7678480 DOI: 10.1136/bmjnph-2018-000004] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Revised: 08/01/2018] [Accepted: 08/02/2018] [Indexed: 12/25/2022] Open
Affiliation(s)
- Olivia M Dong
- Center for Pharmacogenomics and Individualized Therapy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| |
Collapse
|
31
|
Sun CJ, Li L, Li X, Zhang WY, Liu XW. Novel SNPs of WNK1 and AKR1C3 are associated with preeclampsia. Gene 2018; 668:27-32. [PMID: 29777907 DOI: 10.1016/j.gene.2018.05.055] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Accepted: 05/15/2018] [Indexed: 11/26/2022]
Abstract
Preeclampsia is a hypertensive disorder of pregnancy and is one of the most common causes of poor perinatal outcomes. Preeclampsia increases the risk of hypertension in the future. Variants of WNK1 (lysine deficient protein kinase 1), ADRB2 (β2 adrenergic receptor), NEDD4L (ubiquitin-protein ligase NEDD4-like), KLK1 (kallikrein 1) contribute to hypertension, and AKR1C3 (aldo-keto reductase family1 member C3), is associated with preeclampsia. The association of single nucleotide polymorphisms (SNPs) in these five candidate preeclampsia susceptibility genes and the related traits in Chinese individuals were investigated. In this study, 13 SNPs of the five genes were genotyped in 276 preeclampsia patients and 229 age- and area-matched normal pregnancies in women of Chinese Northern Han origin. The 95% confidence interval (CI) and odds ratio (OR) were estimated by binary logistic regression. No obvious linkage disequilibrium or haplotypes were observed among these SNPs. Those with GG genotype and allele G of AKR1C3 (rs10508293) had a decreased risk of preeclampsia (adjusted OR = 3.011, 95% CI = 1.758-5.159, and adjusted OR = 1.745, 95% CI = 1.349-2.257, respectively). The AA genotype and allele A of WNK1 (rs1468326) were significantly associated with an increased risk in preeclampsia (adjusted OR = 2.307, 95% CI = 1.206-3.443, and adjusted OR = 1.663, 95% CI = 1.283-2.157, respectively). The findings indicate that the GG genotype of AKR1C3 rs10508293 is associated with decreased risk for preeclampsia and the AA genotype of WNK1 rs1468326 are related with an increased risk for preeclampsia.
Collapse
Affiliation(s)
- Cheng-Juan Sun
- Department of Obstetrics, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Chaoyang District, Beijing 100026, China
| | - Lin Li
- Central Laboratory, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Chaoyang District, Beijing 100026, China
| | - Xueyan Li
- Department of Obstetrics, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Chaoyang District, Beijing 100026, China
| | - Wei-Yuan Zhang
- Department of Obstetrics, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Chaoyang District, Beijing 100026, China
| | - Xiao-Wei Liu
- Department of Obstetrics, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Chaoyang District, Beijing 100026, China.
| |
Collapse
|
32
|
Seyerle AA, Sitlani CM, Noordam R, Gogarten SM, Li J, Li X, Evans DS, Sun F, Laaksonen MA, Isaacs A, Kristiansson K, Highland HM, Stewart JD, Harris TB, Trompet S, Bis JC, Peloso GM, Brody JA, Broer L, Busch EL, Duan Q, Stilp AM, O'Donnell CJ, Macfarlane PW, Floyd JS, Kors JA, Lin HJ, Li-Gao R, Sofer T, Méndez-Giráldez R, Cummings SR, Heckbert SR, Hofman A, Ford I, Li Y, Launer LJ, Porthan K, Newton-Cheh C, Napier MD, Kerr KF, Reiner AP, Rice KM, Roach J, Buckley BM, Soliman EZ, de Mutsert R, Sotoodehnia N, Uitterlinden AG, North KE, Lee CR, Gudnason V, Stürmer T, Rosendaal FR, Taylor KD, Wiggins KL, Wilson JG, Chen YD, Kaplan RC, Wilhelmsen K, Cupples LA, Salomaa V, van Duijn C, Jukema JW, Liu Y, Mook-Kanamori DO, Lange LA, Vasan RS, Smith AV, Stricker BH, Laurie CC, Rotter JI, Whitsel EA, Psaty BM, Avery CL. Pharmacogenomics study of thiazide diuretics and QT interval in multi-ethnic populations: the cohorts for heart and aging research in genomic epidemiology. THE PHARMACOGENOMICS JOURNAL 2018; 18:215-226. [PMID: 28719597 PMCID: PMC5773415 DOI: 10.1038/tpj.2017.10] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Revised: 01/14/2017] [Accepted: 03/09/2017] [Indexed: 12/23/2022]
Abstract
Thiazide diuretics, commonly used antihypertensives, may cause QT interval (QT) prolongation, a risk factor for highly fatal and difficult to predict ventricular arrhythmias. We examined whether common single-nucleotide polymorphisms (SNPs) modified the association between thiazide use and QT or its component parts (QRS interval, JT interval) by performing ancestry-specific, trans-ethnic and cross-phenotype genome-wide analyses of European (66%), African American (15%) and Hispanic (19%) populations (N=78 199), leveraging longitudinal data, incorporating corrected standard errors to account for underestimation of interaction estimate variances and evaluating evidence for pathway enrichment. Although no loci achieved genome-wide significance (P<5 × 10-8), we found suggestive evidence (P<5 × 10-6) for SNPs modifying the thiazide-QT association at 22 loci, including ion transport loci (for example, NELL1, KCNQ3). The biologic plausibility of our suggestive results and simulations demonstrating modest power to detect interaction effects at genome-wide significant levels indicate that larger studies and innovative statistical methods are warranted in future efforts evaluating thiazide-SNP interactions.
Collapse
Affiliation(s)
- A A Seyerle
- Department of Epidemiology, University of North Carolina, Chapel Hill, NC, USA
- Division of Epidemiology and Community Health, University of Minnesota, Minneapolis, MN, USA
| | - C M Sitlani
- Department of Medicine, University of Washington, Seattle, WA, USA
| | - R Noordam
- Department of Epidemiology, Erasmus MC-University Medical Center Rotterdam, Rotterdam, The Netherlands
- Department of Gerontology and Geriatrics, Leiden University Medical Center, Leiden, The Netherlands
| | - S M Gogarten
- Department of Biostatistics, University of Washington, Seattle, WA, USA
| | - J Li
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Palo Alto, CA, USA
| | - X Li
- Institute for Translational Genomics and Population Sciences, Department of Pediatrics, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - D S Evans
- California Pacific Medical Center Research Institute, San Francisco, CA, USA
| | - F Sun
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - M A Laaksonen
- Department of Health, THL-National Institute for Health and Welfare, Helsinki, Finland
| | - A Isaacs
- Department of Epidemiology, Erasmus MC-University Medical Center Rotterdam, Rotterdam, The Netherlands
- CARIM School of Cardiovascular Diseases, Maastricht Centre for Systems Biology (MaCSBio), and Department of Biochemistry, Maastricht University, Maastricht, The Netherlands
| | - K Kristiansson
- Department of Health, THL-National Institute for Health and Welfare, Helsinki, Finland
| | - H M Highland
- Department of Epidemiology, University of North Carolina, Chapel Hill, NC, USA
| | - J D Stewart
- Department of Epidemiology, University of North Carolina, Chapel Hill, NC, USA
- Carolina Population Center, University of North Carolina, Chapel Hill, NC, USA
| | - T B Harris
- Laboratory of Epidemiology, Demography, and Biometry, National Institute on Aging, Bethesda, MD, USA
| | - S Trompet
- Department of Gerontology and Geriatrics, Leiden University Medical Center, Leiden, The Netherlands
- Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands
| | - J C Bis
- Department of Medicine, University of Washington, Seattle, WA, USA
| | - G M Peloso
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - J A Brody
- Department of Medicine, University of Washington, Seattle, WA, USA
| | - L Broer
- Department of Internal Medicine, Erasmus MC-University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - E L Busch
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Q Duan
- Department of Genetics, University of North Carolina, Chapel Hill, NC, USA
| | - A M Stilp
- Department of Biostatistics, University of Washington, Seattle, WA, USA
| | - C J O'Donnell
- Department of Medicine, Harvard University, Boston, MA, USA
- National Heart, Lung, and Blood Institute Framingham Heart Study, Framingham, MA, USA
- Cardiology Section, Boston Veterans Administration Healthcare, Boston, MA, USA
| | - P W Macfarlane
- Institute of Health and Wellbeing, University of Glasgow, Glasgow, UK
| | - J S Floyd
- Department of Medicine, University of Washington, Seattle, WA, USA
- Department of Epidemiology, University of Washington, Seattle, WA, USA
| | - J A Kors
- Department of Medical Informatics, Erasmus MC-University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - H J Lin
- Institute for Translational Genomics and Population Sciences, Department of Pediatrics, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA, USA
- Division of Medical Genetics, Department of Pediatrics, Harbor-UCLA Medical Center, Torrance, CA, USA
| | - R Li-Gao
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, The Netherlands
| | - T Sofer
- Department of Medicine, University of Washington, Seattle, WA, USA
| | - R Méndez-Giráldez
- Department of Epidemiology, University of North Carolina, Chapel Hill, NC, USA
| | - S R Cummings
- California Pacific Medical Center Research Institute, San Francisco, CA, USA
| | - S R Heckbert
- Department of Epidemiology, University of Washington, Seattle, WA, USA
| | - A Hofman
- Department of Epidemiology, Erasmus MC-University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - I Ford
- Robertson Center for Biostatistics, University of Glasgow, Glasgow, UK
| | - Y Li
- Department of Genetics, University of North Carolina, Chapel Hill, NC, USA
- Department of Biostatistics, University of North Carolina, Chapel Hill, NC, USA
- Department of Computer Science, University of North Carolina, Chapel Hill, NC, USA
| | - L J Launer
- Laboratory of Epidemiology, Demography, and Biometry, National Institute on Aging, Bethesda, MD, USA
| | - K Porthan
- Division of Cardiology, Heart and Lung Center, Helsinki University Central Hospital, Helsinki, Finland
| | - C Newton-Cheh
- Institute of Health and Wellbeing, University of Glasgow, Glasgow, UK
- Center for Human Genetic Research, Massachusetts General Hospital, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute, Cambridge, MA, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
| | - M D Napier
- Department of Epidemiology, University of North Carolina, Chapel Hill, NC, USA
| | - K F Kerr
- Department of Biostatistics, University of Washington, Seattle, WA, USA
| | - A P Reiner
- Department of Epidemiology, University of Washington, Seattle, WA, USA
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - K M Rice
- Department of Biostatistics, University of Washington, Seattle, WA, USA
| | - J Roach
- Research Computing Center, University of North Carolina, Chapel Hill, NC, USA
| | - B M Buckley
- Department of Pharmacology and Therapeutics, University College Cork, Cork, Ireland
| | - E Z Soliman
- Epidemiology Cardiology Research Center (EPICARE), Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - R de Mutsert
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, The Netherlands
| | - N Sotoodehnia
- Department of Epidemiology, University of Washington, Seattle, WA, USA
- Division of Cardiology, University of Washington, Seattle, WA, USA
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
| | - A G Uitterlinden
- Department of Internal Medicine, Erasmus MC-University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - K E North
- Department of Epidemiology, University of North Carolina, Chapel Hill, NC, USA
| | - C R Lee
- Division of Pharmacotherapy and Experimental Therapeutics, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, USA
| | - V Gudnason
- Icelandic Heart Association, Kopavogur, Iceland
- Department of Medicine, University of Iceland, Reykjavik, Iceland
| | - T Stürmer
- Department of Epidemiology, University of North Carolina, Chapel Hill, NC, USA
- Center for Pharmacoepidemiology, University of North Carolina, Chapel Hill, NC, USA
| | - F R Rosendaal
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, The Netherlands
| | - K D Taylor
- Institute for Translational Genomics and Population Sciences, Department of Pediatrics, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - K L Wiggins
- Department of Medicine, University of Washington, Seattle, WA, USA
| | - J G Wilson
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS, USA
| | - Y-Di Chen
- Institute for Translational Genomics and Population Sciences, Department of Pediatrics, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - R C Kaplan
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, USA
| | - K Wilhelmsen
- Department of Genetics, University of North Carolina, Chapel Hill, NC, USA
- The Renaissance Computing Institute, Chapel Hill, NC, USA
| | - L A Cupples
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
- National Heart, Lung, and Blood Institute Framingham Heart Study, Framingham, MA, USA
| | - V Salomaa
- Department of Health, THL-National Institute for Health and Welfare, Helsinki, Finland
| | - C van Duijn
- Department of Epidemiology, Erasmus MC-University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - J W Jukema
- Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands
- Durrer Center for Cardiogenetic Research, Amsterdam, The Netherlands
- Interuniversity Cardiology Institute of the Netherlands, Utrecht, The Netherlands
| | - Y Liu
- Department of Epidemiology and Prevention, Division of Public Health Sciences, Wake Forest University, Winston-Salem, NC, USA
| | - D O Mook-Kanamori
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, The Netherlands
- Department of Public Health and Primary Care, Leiden University Medical Center, Leiden, the Netherlands
- Department of BESC, Epidemiology Section, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - L A Lange
- Department of Genetics, University of North Carolina, Chapel Hill, NC, USA
| | - R S Vasan
- National Heart, Lung, and Blood Institute Framingham Heart Study, Framingham, MA, USA
- Division of Preventive Medicine and Epidemiology, Department of Epidemiology, Boston University School of Medicine, Boston, MA, USA
| | - A V Smith
- Icelandic Heart Association, Kopavogur, Iceland
- Department of Medicine, University of Iceland, Reykjavik, Iceland
| | - B H Stricker
- Department of Epidemiology, Erasmus MC-University Medical Center Rotterdam, Rotterdam, The Netherlands
- Inspectorate of Health Care, Utrecht, The Netherlands
| | - C C Laurie
- Department of Biostatistics, University of Washington, Seattle, WA, USA
| | - J I Rotter
- Institute for Translational Genomics and Population Sciences, Department of Pediatrics, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - E A Whitsel
- Department of Epidemiology, University of North Carolina, Chapel Hill, NC, USA
- Department of Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - B M Psaty
- Department of Medicine, University of Washington, Seattle, WA, USA
- Department of Epidemiology, University of Washington, Seattle, WA, USA
- Department of Health Services, University of Washington, Seattle, WA, USA
- Group Health Research Institute, Group Health Cooperative, Seattle, WA, USA
| | - C L Avery
- Department of Epidemiology, University of North Carolina, Chapel Hill, NC, USA
- Carolina Population Center, University of North Carolina, Chapel Hill, NC, USA
| |
Collapse
|
33
|
Eadon MT, Kanuri SH, Chapman AB. Pharmacogenomic studies of hypertension: paving the way for personalized antihypertensive treatment. EXPERT REVIEW OF PRECISION MEDICINE AND DRUG DEVELOPMENT 2018; 3:33-47. [PMID: 29888336 DOI: 10.1080/23808993.2018.1420419] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Introduction Increasing clinical evidence supports the implementation of genotyping for anti-hypertensive drug dosing and selection. Despite robust evidence gleaned from clinical trials, the translation of genotype guided therapy into clinical practice faces significant challenges. Challenges to implementation include the small effect size of individual variants and the polygenetic nature of antihypertensive drug response, a lack of expert consensus on dosing guidelines even without genetic information, and proper definition of major antihypertensive drug toxicities. Balancing clinical benefit with cost, while overcoming these challenges, remains crucial. Areas covered This review presents the most impactful clinical trials and cohorts which continue to inform and guide future investigation. Variants were selected from among those identified in the Pharmacogenomic Evaluation of Antihypertensive Responses (PEAR), the Genetic Epidemiology of Responses to Antihypertensives study (GERA), the Genetics of Drug Responsiveness in Essential Hypertension (GENRES) study, the SOPHIA study, the Milan Hypertension Pharmacogenomics of hydro-chlorothiazide (MIHYPHCTZ), the Campania Salute Network, the International Verapamil SR Trandolapril Study (INVEST), the Nordic Diltiazem (NORDIL) Study, GenHAT, and others. Expert Commentary The polygenic nature of antihypertensive drug response is a major barrier to clinical implementation. Further studies examining clinical effectiveness are required to support broad-based implementation of genotype-based prescribing in medical practice.
Collapse
Affiliation(s)
- Michael T Eadon
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Sri H Kanuri
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | | |
Collapse
|
34
|
Whole Transcriptome Sequencing Analyses Reveal Molecular Markers of Blood Pressure Response to Thiazide Diuretics. Sci Rep 2017; 7:16068. [PMID: 29167564 PMCID: PMC5700078 DOI: 10.1038/s41598-017-16343-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Accepted: 11/10/2017] [Indexed: 01/13/2023] Open
Abstract
Thiazide diuretics (TD) are commonly prescribed anti-hypertensives worldwide. However, <40% of patients treated with thiazide monotherapy achieve BP control. This study uses whole transcriptome sequencing to identify novel molecular markers associated with BP response to TD. We assessed global RNA expression levels in whole blood samples from 150 participants, representing patients in the upper and lower quartile of BP response to TD from the Pharmacogenomic Evaluation of Antihypertensive Responses (PEAR) (50 whites) and from PEAR-2 (50 whites and 50 blacks). In each study cohort, we performed poly-A RNA-sequencing in baseline samples from 25 responders and 25 non-responders to hydrochlorothiazide (HCTZ) or chlorthalidone. At FDR adjusted p-value < 0.05, 29 genes were differentially expressed in relation to HCTZ or chlorthalidone BP response in whites. For each differentially expressed gene, replication was attempted in the alternate white group and PEAR-2 blacks. CEBPD (meta-analysis p = 1.8 × 10−11) and TSC22D3 (p = 1.9 × 10−9) were differentially expressed in all 3 cohorts, and explain, in aggregate, 21.9% of response variability to TD. This is the first report of the use of transcriptome-wide sequencing data to identify molecular markers of antihypertensive drug response. These findings support CEBPD and TSC22D3 as potential biomarkers of BP response to TD.
Collapse
|
35
|
Personalized medicine-a modern approach for the diagnosis and management of hypertension. Clin Sci (Lond) 2017; 131:2671-2685. [PMID: 29109301 PMCID: PMC5736921 DOI: 10.1042/cs20160407] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2017] [Revised: 09/22/2017] [Accepted: 09/25/2017] [Indexed: 12/15/2022]
Abstract
The main goal of treating hypertension is to reduce blood pressure to physiological levels and thereby prevent risk of cardiovascular disease and hypertension-associated target organ damage. Despite reductions in major risk factors and the availability of a plethora of effective antihypertensive drugs, the control of blood pressure to target values is still poor due to multiple factors including apparent drug resistance and lack of adherence. An explanation for this problem is related to the current reductionist and ‘trial-and-error’ approach in the management of hypertension, as we may oversimplify the complex nature of the disease and not pay enough attention to the heterogeneity of the pathophysiology and clinical presentation of the disorder. Taking into account specific risk factors, genetic phenotype, pharmacokinetic characteristics, and other particular features unique to each patient, would allow a personalized approach to managing the disease. Personalized medicine therefore represents the tailoring of medical approach and treatment to the individual characteristics of each patient and is expected to become the paradigm of future healthcare. The advancement of systems biology research and the rapid development of high-throughput technologies, as well as the characterization of different –omics, have contributed to a shift in modern biological and medical research from traditional hypothesis-driven designs toward data-driven studies and have facilitated the evolution of personalized or precision medicine for chronic diseases such as hypertension.
Collapse
|
36
|
Henshall TL, Manning JA, Alfassy OS, Goel P, Boase NA, Kawabe H, Kumar S. Deletion of Nedd4-2 results in progressive kidney disease in mice. Cell Death Differ 2017; 24:2150-2160. [PMID: 28862701 PMCID: PMC5686353 DOI: 10.1038/cdd.2017.137] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Revised: 07/05/2017] [Accepted: 07/19/2017] [Indexed: 12/30/2022] Open
Abstract
NEDD4-2 (NEDD4L), a ubiquitin protein ligase of the Nedd4 family, is a key regulator of cell surface expression and activity of the amiloride-sensitive epithelial Na+ channel (ENaC). While hypomorphic alleles of Nedd4-2 in mice show salt-sensitive hypertension, complete knockout results in pulmonary distress and perinatal lethality due to increased cell surface levels of ENaC. We now show that Nedd4-2 deficiency in mice also results in an unexpected progressive kidney injury phenotype associated with elevated ENaC and Na+Cl− cotransporter expression, increased Na+ reabsorption, hypertension and markedly reduced levels of aldosterone. The observed nephropathy is characterized by fibrosis, tubule epithelial cell apoptosis, dilated/cystic tubules, elevated expression of kidney injury markers and immune cell infiltration, characteristics reminiscent of human chronic kidney disease. Importantly, we demonstrate that the extent of kidney injury can be partially therapeutically ameliorated in mice with nephron-specific deletions of Nedd4-2 by blocking ENaC with amiloride. These results suggest that increased Na+ reabsorption via ENaC causes kidney injury and establish a novel role of NEDD4-2 in preventing Na+-induced nephropathy. Contrary to some recent reports, our data also indicate that ENaC is the primary in vivo target of NEDD4-2 and that Nedd4-2 deletion is associated with hypertension on a normal Na+ diet. These findings provide further insight into the critical function of NEDD4-2 in renal pathophysiology.
Collapse
Affiliation(s)
- Tanya L Henshall
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, SA 5000, Australia
| | - Jantina A Manning
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, SA 5000, Australia
| | - Omri S Alfassy
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, SA 5000, Australia
| | - Pranay Goel
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, SA 5000, Australia.,School of Medicine, University of Adelaide, Adelaide, SA 5005, Australia
| | - Natasha A Boase
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, SA 5000, Australia
| | - Hiroshi Kawabe
- Max Planck Institute of Experimental Medicine, Department of Molecular Neurobiology, Göttingen 37075, Germany
| | - Sharad Kumar
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, SA 5000, Australia.,School of Medicine, University of Adelaide, Adelaide, SA 5005, Australia
| |
Collapse
|
37
|
Frame AA, Wainford RD. Renal sodium handling and sodium sensitivity. Kidney Res Clin Pract 2017; 36:117-131. [PMID: 28680820 PMCID: PMC5491159 DOI: 10.23876/j.krcp.2017.36.2.117] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Accepted: 12/27/2016] [Indexed: 01/11/2023] Open
Abstract
The pathophysiology of hypertension, which affects over 1 billion individuals worldwide, involves the integration of the actions of multiple organ systems, including the kidney. The kidney, which governs sodium excretion via several mechanisms including pressure natriuresis and the actions of renal sodium transporters, is central to long term blood pressure regulation and the salt sensitivity of blood pressure. The impact of renal sodium handling and the salt sensitivity of blood pressure in health and hypertension is a critical public health issue owing to the excess of dietary salt consumed globally and the significant percentage of the global population exhibiting salt sensitivity. This review highlights recent advances that have provided new insight into the renal handling of sodium and the salt sensitivity of blood pressure, with a focus on genetic, inflammatory, dietary, sympathetic nervous system and oxidative stress mechanisms that influence renal sodium excretion. Increased understanding of the multiple integrated mechanisms that regulate the renal handling of sodium and the salt sensitivity of blood pressure has the potential to identify novel therapeutic targets and refine dietary guidelines designed to treat and prevent hypertension.
Collapse
Affiliation(s)
- Alissa A Frame
- Department of Pharmacology & Experimental Therapeutics and The Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA, USA
| | - Richard D Wainford
- Department of Pharmacology & Experimental Therapeutics and The Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA, USA
| |
Collapse
|
38
|
Zaiou M, El Amri H. Cardiovascular pharmacogenetics: a promise for genomically-guided therapy and personalized medicine. Clin Genet 2017; 91:355-370. [PMID: 27714756 DOI: 10.1111/cge.12881] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Revised: 09/30/2016] [Accepted: 10/03/2016] [Indexed: 12/28/2022]
Abstract
Cardiovascular disease (CVD) is the leading cause of death worldwide. The basic causes of CVD are not fully understood yet. Substantial evidence suggests that genetic predisposition plays a vital role in the physiopathology of this complex disease. Hence, identification of genetic contributors to CVD will likely add diagnostic accuracy and better prediction of an individual's risk. With high-throughput genetics and genomics technology and newer genome-wide study approaches, a number of genetic variations across the human genome were uncovered. Evidence suggests that genetic defects could influence CVD development and inter-individual responses to widely used cardiovascular drugs like clopidogrel, aspirin, warfarin, and statins, and therefore, they may be integrated into clinical practice. If clinically validated, better understanding of these genetic variations may provide new opportunities for personalized diagnostic, pharmacogenetic-based drug selection and best treatment in personalized medicine. However, numerous gaps remain unsolved due to the lack of underlying pathological mechanisms for how genetic predisposition could contribute to CVD. This review provides an overview of the extraordinary scientific progress in our understanding of genetic and genomic basis of CVD as well as the development of relevant genetic biomarkers for this disease. Some of the actual limitations to the promise of these markers and their translation for the benefit of patients will be discussed.
Collapse
Affiliation(s)
- M Zaiou
- Faculté de Pharmacie, Université de Lorraine, Nancy, France
| | - H El Amri
- Laboratoire de Génétique de la Gendarmerie Royale, Avenue Ibn Sina, Rabat, Maroc
| |
Collapse
|
39
|
Salvi E, Wang Z, Rizzi F, Gong Y, McDonough CW, Padmanabhan S, Hiltunen TP, Lanzani C, Zaninello R, Chittani M, Bailey KR, Sarin AP, Barcella M, Melander O, Chapman AB, Manunta P, Kontula KK, Glorioso N, Cusi D, Dominiczak AF, Johnson JA, Barlassina C, Boerwinkle E, Cooper-DeHoff RM, Turner ST. Genome-Wide and Gene-Based Meta-Analyses Identify Novel Loci Influencing Blood Pressure Response to Hydrochlorothiazide. Hypertension 2017; 69:51-59. [PMID: 27802415 PMCID: PMC5145728 DOI: 10.1161/hypertensionaha.116.08267] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Revised: 08/26/2016] [Accepted: 10/07/2016] [Indexed: 12/28/2022]
Abstract
This study aimed to identify novel loci influencing the antihypertensive response to hydrochlorothiazide monotherapy. A genome-wide meta-analysis of blood pressure (BP) response to hydrochlorothiazide was performed in 1739 white hypertensives from 6 clinical trials within the International Consortium for Antihypertensive Pharmacogenomics Studies, making it the largest study to date of its kind. No signals reached genome-wide significance (P<5×10-8), and the suggestive regions (P<10-5) were cross-validated in 2 black cohorts treated with hydrochlorothiazide. In addition, a gene-based analysis was performed on candidate genes with previous evidence of involvement in diuretic response, in BP regulation, or in hypertension susceptibility. Using the genome-wide meta-analysis approach, with validation in blacks, we identified 2 suggestive regulatory regions linked to gap junction protein α1 gene (GJA1) and forkhead box A1 gene (FOXA1), relevant for cardiovascular and kidney function. With the gene-based approach, we identified hydroxy-delta-5-steroid dehydrogenase, 3 β- and steroid δ-isomerase 1 gene (HSD3B1) as significantly associated with BP response (P<2.28×10-4 ). HSD3B1 encodes the 3β-hydroxysteroid dehydrogenase enzyme and plays a crucial role in the biosynthesis of aldosterone and endogenous ouabain. By amassing all of the available pharmacogenomic studies of BP response to hydrochlorothiazide, and using 2 different analytic approaches, we identified 3 novel loci influencing BP response to hydrochlorothiazide. The gene-based analysis, never before applied to pharmacogenomics of antihypertensive drugs to our knowledge, provided a powerful strategy to identify a locus of interest, which was not identified in the genome-wide meta-analysis because of high allelic heterogeneity. These data pave the way for future investigations on new pathways and drug targets to enhance the current understanding of personalized antihypertensive treatment.
Collapse
Affiliation(s)
- Erika Salvi
- From the Department of Health Sciences, University of Milan, Italy (E.S., F.R., M.C., M.B., C.B.); Human Genetics and Institute of Molecular Medicine, University of Texas Health Science Center, Houston (Z.W., E.B.); Department of Pharmacotherapy and Translational Research, Center for Pharmacogenomics, College of Pharmacy (Y.G., C.W.M., J.A.J., R.M.C.-D.) and Division of Cardiovascular Medicine, Department of Medicine (J.A.J., R.M.C.-D.), University of Florida, Gainesville; Institute of Cardiovascular and Medical Sciences, College of Medical Veterinary and Life Sciences, University of Glasgow, United Kingdom (S.P., A.F.D.); Department of Medicine, University of Helsinki and Helsinki University Hospital, Finland (T.P.H., K.K.K.); Nephrology and Dialysis and Hypertension Unit, San Raffaele Scientific Institute, Università Vita Salute San Raffaele, Milano, Italy (C.L., P.M.); Hypertension and Related Disease Centre, AOU-University of Sassari, Italy (R.Z., N.G.); Division of Biomedical Statistics and Informatics, Department of Health Sciences Research (K.R.B.) and Division of Nephrology and Hypertension, Department of Internal Medicine (S.T.T.), Mayo Clinic, Rochester, Minnesota; Institute for Molecular Medicine Finland FIMM, University of Helsinki, Finland (A.-P.S); Department of Clinical Sciences, Lund University, Malmö, Sweden (O.M.); Section of Nephrology, Department of Medicine, University of Chicago, Illinois (A.B.C.); Institute of Biomedical Technologies, National Research Centre of Italy, Segrate, Milan, Italy (D.C.); and Sanipedia srl, Bresso, Italy (D.C.).
| | - Zhiying Wang
- From the Department of Health Sciences, University of Milan, Italy (E.S., F.R., M.C., M.B., C.B.); Human Genetics and Institute of Molecular Medicine, University of Texas Health Science Center, Houston (Z.W., E.B.); Department of Pharmacotherapy and Translational Research, Center for Pharmacogenomics, College of Pharmacy (Y.G., C.W.M., J.A.J., R.M.C.-D.) and Division of Cardiovascular Medicine, Department of Medicine (J.A.J., R.M.C.-D.), University of Florida, Gainesville; Institute of Cardiovascular and Medical Sciences, College of Medical Veterinary and Life Sciences, University of Glasgow, United Kingdom (S.P., A.F.D.); Department of Medicine, University of Helsinki and Helsinki University Hospital, Finland (T.P.H., K.K.K.); Nephrology and Dialysis and Hypertension Unit, San Raffaele Scientific Institute, Università Vita Salute San Raffaele, Milano, Italy (C.L., P.M.); Hypertension and Related Disease Centre, AOU-University of Sassari, Italy (R.Z., N.G.); Division of Biomedical Statistics and Informatics, Department of Health Sciences Research (K.R.B.) and Division of Nephrology and Hypertension, Department of Internal Medicine (S.T.T.), Mayo Clinic, Rochester, Minnesota; Institute for Molecular Medicine Finland FIMM, University of Helsinki, Finland (A.-P.S); Department of Clinical Sciences, Lund University, Malmö, Sweden (O.M.); Section of Nephrology, Department of Medicine, University of Chicago, Illinois (A.B.C.); Institute of Biomedical Technologies, National Research Centre of Italy, Segrate, Milan, Italy (D.C.); and Sanipedia srl, Bresso, Italy (D.C.)
| | - Federica Rizzi
- From the Department of Health Sciences, University of Milan, Italy (E.S., F.R., M.C., M.B., C.B.); Human Genetics and Institute of Molecular Medicine, University of Texas Health Science Center, Houston (Z.W., E.B.); Department of Pharmacotherapy and Translational Research, Center for Pharmacogenomics, College of Pharmacy (Y.G., C.W.M., J.A.J., R.M.C.-D.) and Division of Cardiovascular Medicine, Department of Medicine (J.A.J., R.M.C.-D.), University of Florida, Gainesville; Institute of Cardiovascular and Medical Sciences, College of Medical Veterinary and Life Sciences, University of Glasgow, United Kingdom (S.P., A.F.D.); Department of Medicine, University of Helsinki and Helsinki University Hospital, Finland (T.P.H., K.K.K.); Nephrology and Dialysis and Hypertension Unit, San Raffaele Scientific Institute, Università Vita Salute San Raffaele, Milano, Italy (C.L., P.M.); Hypertension and Related Disease Centre, AOU-University of Sassari, Italy (R.Z., N.G.); Division of Biomedical Statistics and Informatics, Department of Health Sciences Research (K.R.B.) and Division of Nephrology and Hypertension, Department of Internal Medicine (S.T.T.), Mayo Clinic, Rochester, Minnesota; Institute for Molecular Medicine Finland FIMM, University of Helsinki, Finland (A.-P.S); Department of Clinical Sciences, Lund University, Malmö, Sweden (O.M.); Section of Nephrology, Department of Medicine, University of Chicago, Illinois (A.B.C.); Institute of Biomedical Technologies, National Research Centre of Italy, Segrate, Milan, Italy (D.C.); and Sanipedia srl, Bresso, Italy (D.C.)
| | - Yan Gong
- From the Department of Health Sciences, University of Milan, Italy (E.S., F.R., M.C., M.B., C.B.); Human Genetics and Institute of Molecular Medicine, University of Texas Health Science Center, Houston (Z.W., E.B.); Department of Pharmacotherapy and Translational Research, Center for Pharmacogenomics, College of Pharmacy (Y.G., C.W.M., J.A.J., R.M.C.-D.) and Division of Cardiovascular Medicine, Department of Medicine (J.A.J., R.M.C.-D.), University of Florida, Gainesville; Institute of Cardiovascular and Medical Sciences, College of Medical Veterinary and Life Sciences, University of Glasgow, United Kingdom (S.P., A.F.D.); Department of Medicine, University of Helsinki and Helsinki University Hospital, Finland (T.P.H., K.K.K.); Nephrology and Dialysis and Hypertension Unit, San Raffaele Scientific Institute, Università Vita Salute San Raffaele, Milano, Italy (C.L., P.M.); Hypertension and Related Disease Centre, AOU-University of Sassari, Italy (R.Z., N.G.); Division of Biomedical Statistics and Informatics, Department of Health Sciences Research (K.R.B.) and Division of Nephrology and Hypertension, Department of Internal Medicine (S.T.T.), Mayo Clinic, Rochester, Minnesota; Institute for Molecular Medicine Finland FIMM, University of Helsinki, Finland (A.-P.S); Department of Clinical Sciences, Lund University, Malmö, Sweden (O.M.); Section of Nephrology, Department of Medicine, University of Chicago, Illinois (A.B.C.); Institute of Biomedical Technologies, National Research Centre of Italy, Segrate, Milan, Italy (D.C.); and Sanipedia srl, Bresso, Italy (D.C.)
| | - Caitrin W McDonough
- From the Department of Health Sciences, University of Milan, Italy (E.S., F.R., M.C., M.B., C.B.); Human Genetics and Institute of Molecular Medicine, University of Texas Health Science Center, Houston (Z.W., E.B.); Department of Pharmacotherapy and Translational Research, Center for Pharmacogenomics, College of Pharmacy (Y.G., C.W.M., J.A.J., R.M.C.-D.) and Division of Cardiovascular Medicine, Department of Medicine (J.A.J., R.M.C.-D.), University of Florida, Gainesville; Institute of Cardiovascular and Medical Sciences, College of Medical Veterinary and Life Sciences, University of Glasgow, United Kingdom (S.P., A.F.D.); Department of Medicine, University of Helsinki and Helsinki University Hospital, Finland (T.P.H., K.K.K.); Nephrology and Dialysis and Hypertension Unit, San Raffaele Scientific Institute, Università Vita Salute San Raffaele, Milano, Italy (C.L., P.M.); Hypertension and Related Disease Centre, AOU-University of Sassari, Italy (R.Z., N.G.); Division of Biomedical Statistics and Informatics, Department of Health Sciences Research (K.R.B.) and Division of Nephrology and Hypertension, Department of Internal Medicine (S.T.T.), Mayo Clinic, Rochester, Minnesota; Institute for Molecular Medicine Finland FIMM, University of Helsinki, Finland (A.-P.S); Department of Clinical Sciences, Lund University, Malmö, Sweden (O.M.); Section of Nephrology, Department of Medicine, University of Chicago, Illinois (A.B.C.); Institute of Biomedical Technologies, National Research Centre of Italy, Segrate, Milan, Italy (D.C.); and Sanipedia srl, Bresso, Italy (D.C.)
| | - Sandosh Padmanabhan
- From the Department of Health Sciences, University of Milan, Italy (E.S., F.R., M.C., M.B., C.B.); Human Genetics and Institute of Molecular Medicine, University of Texas Health Science Center, Houston (Z.W., E.B.); Department of Pharmacotherapy and Translational Research, Center for Pharmacogenomics, College of Pharmacy (Y.G., C.W.M., J.A.J., R.M.C.-D.) and Division of Cardiovascular Medicine, Department of Medicine (J.A.J., R.M.C.-D.), University of Florida, Gainesville; Institute of Cardiovascular and Medical Sciences, College of Medical Veterinary and Life Sciences, University of Glasgow, United Kingdom (S.P., A.F.D.); Department of Medicine, University of Helsinki and Helsinki University Hospital, Finland (T.P.H., K.K.K.); Nephrology and Dialysis and Hypertension Unit, San Raffaele Scientific Institute, Università Vita Salute San Raffaele, Milano, Italy (C.L., P.M.); Hypertension and Related Disease Centre, AOU-University of Sassari, Italy (R.Z., N.G.); Division of Biomedical Statistics and Informatics, Department of Health Sciences Research (K.R.B.) and Division of Nephrology and Hypertension, Department of Internal Medicine (S.T.T.), Mayo Clinic, Rochester, Minnesota; Institute for Molecular Medicine Finland FIMM, University of Helsinki, Finland (A.-P.S); Department of Clinical Sciences, Lund University, Malmö, Sweden (O.M.); Section of Nephrology, Department of Medicine, University of Chicago, Illinois (A.B.C.); Institute of Biomedical Technologies, National Research Centre of Italy, Segrate, Milan, Italy (D.C.); and Sanipedia srl, Bresso, Italy (D.C.)
| | - Timo P Hiltunen
- From the Department of Health Sciences, University of Milan, Italy (E.S., F.R., M.C., M.B., C.B.); Human Genetics and Institute of Molecular Medicine, University of Texas Health Science Center, Houston (Z.W., E.B.); Department of Pharmacotherapy and Translational Research, Center for Pharmacogenomics, College of Pharmacy (Y.G., C.W.M., J.A.J., R.M.C.-D.) and Division of Cardiovascular Medicine, Department of Medicine (J.A.J., R.M.C.-D.), University of Florida, Gainesville; Institute of Cardiovascular and Medical Sciences, College of Medical Veterinary and Life Sciences, University of Glasgow, United Kingdom (S.P., A.F.D.); Department of Medicine, University of Helsinki and Helsinki University Hospital, Finland (T.P.H., K.K.K.); Nephrology and Dialysis and Hypertension Unit, San Raffaele Scientific Institute, Università Vita Salute San Raffaele, Milano, Italy (C.L., P.M.); Hypertension and Related Disease Centre, AOU-University of Sassari, Italy (R.Z., N.G.); Division of Biomedical Statistics and Informatics, Department of Health Sciences Research (K.R.B.) and Division of Nephrology and Hypertension, Department of Internal Medicine (S.T.T.), Mayo Clinic, Rochester, Minnesota; Institute for Molecular Medicine Finland FIMM, University of Helsinki, Finland (A.-P.S); Department of Clinical Sciences, Lund University, Malmö, Sweden (O.M.); Section of Nephrology, Department of Medicine, University of Chicago, Illinois (A.B.C.); Institute of Biomedical Technologies, National Research Centre of Italy, Segrate, Milan, Italy (D.C.); and Sanipedia srl, Bresso, Italy (D.C.)
| | - Chiara Lanzani
- From the Department of Health Sciences, University of Milan, Italy (E.S., F.R., M.C., M.B., C.B.); Human Genetics and Institute of Molecular Medicine, University of Texas Health Science Center, Houston (Z.W., E.B.); Department of Pharmacotherapy and Translational Research, Center for Pharmacogenomics, College of Pharmacy (Y.G., C.W.M., J.A.J., R.M.C.-D.) and Division of Cardiovascular Medicine, Department of Medicine (J.A.J., R.M.C.-D.), University of Florida, Gainesville; Institute of Cardiovascular and Medical Sciences, College of Medical Veterinary and Life Sciences, University of Glasgow, United Kingdom (S.P., A.F.D.); Department of Medicine, University of Helsinki and Helsinki University Hospital, Finland (T.P.H., K.K.K.); Nephrology and Dialysis and Hypertension Unit, San Raffaele Scientific Institute, Università Vita Salute San Raffaele, Milano, Italy (C.L., P.M.); Hypertension and Related Disease Centre, AOU-University of Sassari, Italy (R.Z., N.G.); Division of Biomedical Statistics and Informatics, Department of Health Sciences Research (K.R.B.) and Division of Nephrology and Hypertension, Department of Internal Medicine (S.T.T.), Mayo Clinic, Rochester, Minnesota; Institute for Molecular Medicine Finland FIMM, University of Helsinki, Finland (A.-P.S); Department of Clinical Sciences, Lund University, Malmö, Sweden (O.M.); Section of Nephrology, Department of Medicine, University of Chicago, Illinois (A.B.C.); Institute of Biomedical Technologies, National Research Centre of Italy, Segrate, Milan, Italy (D.C.); and Sanipedia srl, Bresso, Italy (D.C.)
| | - Roberta Zaninello
- From the Department of Health Sciences, University of Milan, Italy (E.S., F.R., M.C., M.B., C.B.); Human Genetics and Institute of Molecular Medicine, University of Texas Health Science Center, Houston (Z.W., E.B.); Department of Pharmacotherapy and Translational Research, Center for Pharmacogenomics, College of Pharmacy (Y.G., C.W.M., J.A.J., R.M.C.-D.) and Division of Cardiovascular Medicine, Department of Medicine (J.A.J., R.M.C.-D.), University of Florida, Gainesville; Institute of Cardiovascular and Medical Sciences, College of Medical Veterinary and Life Sciences, University of Glasgow, United Kingdom (S.P., A.F.D.); Department of Medicine, University of Helsinki and Helsinki University Hospital, Finland (T.P.H., K.K.K.); Nephrology and Dialysis and Hypertension Unit, San Raffaele Scientific Institute, Università Vita Salute San Raffaele, Milano, Italy (C.L., P.M.); Hypertension and Related Disease Centre, AOU-University of Sassari, Italy (R.Z., N.G.); Division of Biomedical Statistics and Informatics, Department of Health Sciences Research (K.R.B.) and Division of Nephrology and Hypertension, Department of Internal Medicine (S.T.T.), Mayo Clinic, Rochester, Minnesota; Institute for Molecular Medicine Finland FIMM, University of Helsinki, Finland (A.-P.S); Department of Clinical Sciences, Lund University, Malmö, Sweden (O.M.); Section of Nephrology, Department of Medicine, University of Chicago, Illinois (A.B.C.); Institute of Biomedical Technologies, National Research Centre of Italy, Segrate, Milan, Italy (D.C.); and Sanipedia srl, Bresso, Italy (D.C.)
| | - Martina Chittani
- From the Department of Health Sciences, University of Milan, Italy (E.S., F.R., M.C., M.B., C.B.); Human Genetics and Institute of Molecular Medicine, University of Texas Health Science Center, Houston (Z.W., E.B.); Department of Pharmacotherapy and Translational Research, Center for Pharmacogenomics, College of Pharmacy (Y.G., C.W.M., J.A.J., R.M.C.-D.) and Division of Cardiovascular Medicine, Department of Medicine (J.A.J., R.M.C.-D.), University of Florida, Gainesville; Institute of Cardiovascular and Medical Sciences, College of Medical Veterinary and Life Sciences, University of Glasgow, United Kingdom (S.P., A.F.D.); Department of Medicine, University of Helsinki and Helsinki University Hospital, Finland (T.P.H., K.K.K.); Nephrology and Dialysis and Hypertension Unit, San Raffaele Scientific Institute, Università Vita Salute San Raffaele, Milano, Italy (C.L., P.M.); Hypertension and Related Disease Centre, AOU-University of Sassari, Italy (R.Z., N.G.); Division of Biomedical Statistics and Informatics, Department of Health Sciences Research (K.R.B.) and Division of Nephrology and Hypertension, Department of Internal Medicine (S.T.T.), Mayo Clinic, Rochester, Minnesota; Institute for Molecular Medicine Finland FIMM, University of Helsinki, Finland (A.-P.S); Department of Clinical Sciences, Lund University, Malmö, Sweden (O.M.); Section of Nephrology, Department of Medicine, University of Chicago, Illinois (A.B.C.); Institute of Biomedical Technologies, National Research Centre of Italy, Segrate, Milan, Italy (D.C.); and Sanipedia srl, Bresso, Italy (D.C.)
| | - Kent R Bailey
- From the Department of Health Sciences, University of Milan, Italy (E.S., F.R., M.C., M.B., C.B.); Human Genetics and Institute of Molecular Medicine, University of Texas Health Science Center, Houston (Z.W., E.B.); Department of Pharmacotherapy and Translational Research, Center for Pharmacogenomics, College of Pharmacy (Y.G., C.W.M., J.A.J., R.M.C.-D.) and Division of Cardiovascular Medicine, Department of Medicine (J.A.J., R.M.C.-D.), University of Florida, Gainesville; Institute of Cardiovascular and Medical Sciences, College of Medical Veterinary and Life Sciences, University of Glasgow, United Kingdom (S.P., A.F.D.); Department of Medicine, University of Helsinki and Helsinki University Hospital, Finland (T.P.H., K.K.K.); Nephrology and Dialysis and Hypertension Unit, San Raffaele Scientific Institute, Università Vita Salute San Raffaele, Milano, Italy (C.L., P.M.); Hypertension and Related Disease Centre, AOU-University of Sassari, Italy (R.Z., N.G.); Division of Biomedical Statistics and Informatics, Department of Health Sciences Research (K.R.B.) and Division of Nephrology and Hypertension, Department of Internal Medicine (S.T.T.), Mayo Clinic, Rochester, Minnesota; Institute for Molecular Medicine Finland FIMM, University of Helsinki, Finland (A.-P.S); Department of Clinical Sciences, Lund University, Malmö, Sweden (O.M.); Section of Nephrology, Department of Medicine, University of Chicago, Illinois (A.B.C.); Institute of Biomedical Technologies, National Research Centre of Italy, Segrate, Milan, Italy (D.C.); and Sanipedia srl, Bresso, Italy (D.C.)
| | - Antti-Pekka Sarin
- From the Department of Health Sciences, University of Milan, Italy (E.S., F.R., M.C., M.B., C.B.); Human Genetics and Institute of Molecular Medicine, University of Texas Health Science Center, Houston (Z.W., E.B.); Department of Pharmacotherapy and Translational Research, Center for Pharmacogenomics, College of Pharmacy (Y.G., C.W.M., J.A.J., R.M.C.-D.) and Division of Cardiovascular Medicine, Department of Medicine (J.A.J., R.M.C.-D.), University of Florida, Gainesville; Institute of Cardiovascular and Medical Sciences, College of Medical Veterinary and Life Sciences, University of Glasgow, United Kingdom (S.P., A.F.D.); Department of Medicine, University of Helsinki and Helsinki University Hospital, Finland (T.P.H., K.K.K.); Nephrology and Dialysis and Hypertension Unit, San Raffaele Scientific Institute, Università Vita Salute San Raffaele, Milano, Italy (C.L., P.M.); Hypertension and Related Disease Centre, AOU-University of Sassari, Italy (R.Z., N.G.); Division of Biomedical Statistics and Informatics, Department of Health Sciences Research (K.R.B.) and Division of Nephrology and Hypertension, Department of Internal Medicine (S.T.T.), Mayo Clinic, Rochester, Minnesota; Institute for Molecular Medicine Finland FIMM, University of Helsinki, Finland (A.-P.S); Department of Clinical Sciences, Lund University, Malmö, Sweden (O.M.); Section of Nephrology, Department of Medicine, University of Chicago, Illinois (A.B.C.); Institute of Biomedical Technologies, National Research Centre of Italy, Segrate, Milan, Italy (D.C.); and Sanipedia srl, Bresso, Italy (D.C.)
| | - Matteo Barcella
- From the Department of Health Sciences, University of Milan, Italy (E.S., F.R., M.C., M.B., C.B.); Human Genetics and Institute of Molecular Medicine, University of Texas Health Science Center, Houston (Z.W., E.B.); Department of Pharmacotherapy and Translational Research, Center for Pharmacogenomics, College of Pharmacy (Y.G., C.W.M., J.A.J., R.M.C.-D.) and Division of Cardiovascular Medicine, Department of Medicine (J.A.J., R.M.C.-D.), University of Florida, Gainesville; Institute of Cardiovascular and Medical Sciences, College of Medical Veterinary and Life Sciences, University of Glasgow, United Kingdom (S.P., A.F.D.); Department of Medicine, University of Helsinki and Helsinki University Hospital, Finland (T.P.H., K.K.K.); Nephrology and Dialysis and Hypertension Unit, San Raffaele Scientific Institute, Università Vita Salute San Raffaele, Milano, Italy (C.L., P.M.); Hypertension and Related Disease Centre, AOU-University of Sassari, Italy (R.Z., N.G.); Division of Biomedical Statistics and Informatics, Department of Health Sciences Research (K.R.B.) and Division of Nephrology and Hypertension, Department of Internal Medicine (S.T.T.), Mayo Clinic, Rochester, Minnesota; Institute for Molecular Medicine Finland FIMM, University of Helsinki, Finland (A.-P.S); Department of Clinical Sciences, Lund University, Malmö, Sweden (O.M.); Section of Nephrology, Department of Medicine, University of Chicago, Illinois (A.B.C.); Institute of Biomedical Technologies, National Research Centre of Italy, Segrate, Milan, Italy (D.C.); and Sanipedia srl, Bresso, Italy (D.C.)
| | - Olle Melander
- From the Department of Health Sciences, University of Milan, Italy (E.S., F.R., M.C., M.B., C.B.); Human Genetics and Institute of Molecular Medicine, University of Texas Health Science Center, Houston (Z.W., E.B.); Department of Pharmacotherapy and Translational Research, Center for Pharmacogenomics, College of Pharmacy (Y.G., C.W.M., J.A.J., R.M.C.-D.) and Division of Cardiovascular Medicine, Department of Medicine (J.A.J., R.M.C.-D.), University of Florida, Gainesville; Institute of Cardiovascular and Medical Sciences, College of Medical Veterinary and Life Sciences, University of Glasgow, United Kingdom (S.P., A.F.D.); Department of Medicine, University of Helsinki and Helsinki University Hospital, Finland (T.P.H., K.K.K.); Nephrology and Dialysis and Hypertension Unit, San Raffaele Scientific Institute, Università Vita Salute San Raffaele, Milano, Italy (C.L., P.M.); Hypertension and Related Disease Centre, AOU-University of Sassari, Italy (R.Z., N.G.); Division of Biomedical Statistics and Informatics, Department of Health Sciences Research (K.R.B.) and Division of Nephrology and Hypertension, Department of Internal Medicine (S.T.T.), Mayo Clinic, Rochester, Minnesota; Institute for Molecular Medicine Finland FIMM, University of Helsinki, Finland (A.-P.S); Department of Clinical Sciences, Lund University, Malmö, Sweden (O.M.); Section of Nephrology, Department of Medicine, University of Chicago, Illinois (A.B.C.); Institute of Biomedical Technologies, National Research Centre of Italy, Segrate, Milan, Italy (D.C.); and Sanipedia srl, Bresso, Italy (D.C.)
| | - Arlene B Chapman
- From the Department of Health Sciences, University of Milan, Italy (E.S., F.R., M.C., M.B., C.B.); Human Genetics and Institute of Molecular Medicine, University of Texas Health Science Center, Houston (Z.W., E.B.); Department of Pharmacotherapy and Translational Research, Center for Pharmacogenomics, College of Pharmacy (Y.G., C.W.M., J.A.J., R.M.C.-D.) and Division of Cardiovascular Medicine, Department of Medicine (J.A.J., R.M.C.-D.), University of Florida, Gainesville; Institute of Cardiovascular and Medical Sciences, College of Medical Veterinary and Life Sciences, University of Glasgow, United Kingdom (S.P., A.F.D.); Department of Medicine, University of Helsinki and Helsinki University Hospital, Finland (T.P.H., K.K.K.); Nephrology and Dialysis and Hypertension Unit, San Raffaele Scientific Institute, Università Vita Salute San Raffaele, Milano, Italy (C.L., P.M.); Hypertension and Related Disease Centre, AOU-University of Sassari, Italy (R.Z., N.G.); Division of Biomedical Statistics and Informatics, Department of Health Sciences Research (K.R.B.) and Division of Nephrology and Hypertension, Department of Internal Medicine (S.T.T.), Mayo Clinic, Rochester, Minnesota; Institute for Molecular Medicine Finland FIMM, University of Helsinki, Finland (A.-P.S); Department of Clinical Sciences, Lund University, Malmö, Sweden (O.M.); Section of Nephrology, Department of Medicine, University of Chicago, Illinois (A.B.C.); Institute of Biomedical Technologies, National Research Centre of Italy, Segrate, Milan, Italy (D.C.); and Sanipedia srl, Bresso, Italy (D.C.)
| | - Paolo Manunta
- From the Department of Health Sciences, University of Milan, Italy (E.S., F.R., M.C., M.B., C.B.); Human Genetics and Institute of Molecular Medicine, University of Texas Health Science Center, Houston (Z.W., E.B.); Department of Pharmacotherapy and Translational Research, Center for Pharmacogenomics, College of Pharmacy (Y.G., C.W.M., J.A.J., R.M.C.-D.) and Division of Cardiovascular Medicine, Department of Medicine (J.A.J., R.M.C.-D.), University of Florida, Gainesville; Institute of Cardiovascular and Medical Sciences, College of Medical Veterinary and Life Sciences, University of Glasgow, United Kingdom (S.P., A.F.D.); Department of Medicine, University of Helsinki and Helsinki University Hospital, Finland (T.P.H., K.K.K.); Nephrology and Dialysis and Hypertension Unit, San Raffaele Scientific Institute, Università Vita Salute San Raffaele, Milano, Italy (C.L., P.M.); Hypertension and Related Disease Centre, AOU-University of Sassari, Italy (R.Z., N.G.); Division of Biomedical Statistics and Informatics, Department of Health Sciences Research (K.R.B.) and Division of Nephrology and Hypertension, Department of Internal Medicine (S.T.T.), Mayo Clinic, Rochester, Minnesota; Institute for Molecular Medicine Finland FIMM, University of Helsinki, Finland (A.-P.S); Department of Clinical Sciences, Lund University, Malmö, Sweden (O.M.); Section of Nephrology, Department of Medicine, University of Chicago, Illinois (A.B.C.); Institute of Biomedical Technologies, National Research Centre of Italy, Segrate, Milan, Italy (D.C.); and Sanipedia srl, Bresso, Italy (D.C.)
| | - Kimmo K Kontula
- From the Department of Health Sciences, University of Milan, Italy (E.S., F.R., M.C., M.B., C.B.); Human Genetics and Institute of Molecular Medicine, University of Texas Health Science Center, Houston (Z.W., E.B.); Department of Pharmacotherapy and Translational Research, Center for Pharmacogenomics, College of Pharmacy (Y.G., C.W.M., J.A.J., R.M.C.-D.) and Division of Cardiovascular Medicine, Department of Medicine (J.A.J., R.M.C.-D.), University of Florida, Gainesville; Institute of Cardiovascular and Medical Sciences, College of Medical Veterinary and Life Sciences, University of Glasgow, United Kingdom (S.P., A.F.D.); Department of Medicine, University of Helsinki and Helsinki University Hospital, Finland (T.P.H., K.K.K.); Nephrology and Dialysis and Hypertension Unit, San Raffaele Scientific Institute, Università Vita Salute San Raffaele, Milano, Italy (C.L., P.M.); Hypertension and Related Disease Centre, AOU-University of Sassari, Italy (R.Z., N.G.); Division of Biomedical Statistics and Informatics, Department of Health Sciences Research (K.R.B.) and Division of Nephrology and Hypertension, Department of Internal Medicine (S.T.T.), Mayo Clinic, Rochester, Minnesota; Institute for Molecular Medicine Finland FIMM, University of Helsinki, Finland (A.-P.S); Department of Clinical Sciences, Lund University, Malmö, Sweden (O.M.); Section of Nephrology, Department of Medicine, University of Chicago, Illinois (A.B.C.); Institute of Biomedical Technologies, National Research Centre of Italy, Segrate, Milan, Italy (D.C.); and Sanipedia srl, Bresso, Italy (D.C.)
| | - Nicola Glorioso
- From the Department of Health Sciences, University of Milan, Italy (E.S., F.R., M.C., M.B., C.B.); Human Genetics and Institute of Molecular Medicine, University of Texas Health Science Center, Houston (Z.W., E.B.); Department of Pharmacotherapy and Translational Research, Center for Pharmacogenomics, College of Pharmacy (Y.G., C.W.M., J.A.J., R.M.C.-D.) and Division of Cardiovascular Medicine, Department of Medicine (J.A.J., R.M.C.-D.), University of Florida, Gainesville; Institute of Cardiovascular and Medical Sciences, College of Medical Veterinary and Life Sciences, University of Glasgow, United Kingdom (S.P., A.F.D.); Department of Medicine, University of Helsinki and Helsinki University Hospital, Finland (T.P.H., K.K.K.); Nephrology and Dialysis and Hypertension Unit, San Raffaele Scientific Institute, Università Vita Salute San Raffaele, Milano, Italy (C.L., P.M.); Hypertension and Related Disease Centre, AOU-University of Sassari, Italy (R.Z., N.G.); Division of Biomedical Statistics and Informatics, Department of Health Sciences Research (K.R.B.) and Division of Nephrology and Hypertension, Department of Internal Medicine (S.T.T.), Mayo Clinic, Rochester, Minnesota; Institute for Molecular Medicine Finland FIMM, University of Helsinki, Finland (A.-P.S); Department of Clinical Sciences, Lund University, Malmö, Sweden (O.M.); Section of Nephrology, Department of Medicine, University of Chicago, Illinois (A.B.C.); Institute of Biomedical Technologies, National Research Centre of Italy, Segrate, Milan, Italy (D.C.); and Sanipedia srl, Bresso, Italy (D.C.)
| | - Daniele Cusi
- From the Department of Health Sciences, University of Milan, Italy (E.S., F.R., M.C., M.B., C.B.); Human Genetics and Institute of Molecular Medicine, University of Texas Health Science Center, Houston (Z.W., E.B.); Department of Pharmacotherapy and Translational Research, Center for Pharmacogenomics, College of Pharmacy (Y.G., C.W.M., J.A.J., R.M.C.-D.) and Division of Cardiovascular Medicine, Department of Medicine (J.A.J., R.M.C.-D.), University of Florida, Gainesville; Institute of Cardiovascular and Medical Sciences, College of Medical Veterinary and Life Sciences, University of Glasgow, United Kingdom (S.P., A.F.D.); Department of Medicine, University of Helsinki and Helsinki University Hospital, Finland (T.P.H., K.K.K.); Nephrology and Dialysis and Hypertension Unit, San Raffaele Scientific Institute, Università Vita Salute San Raffaele, Milano, Italy (C.L., P.M.); Hypertension and Related Disease Centre, AOU-University of Sassari, Italy (R.Z., N.G.); Division of Biomedical Statistics and Informatics, Department of Health Sciences Research (K.R.B.) and Division of Nephrology and Hypertension, Department of Internal Medicine (S.T.T.), Mayo Clinic, Rochester, Minnesota; Institute for Molecular Medicine Finland FIMM, University of Helsinki, Finland (A.-P.S); Department of Clinical Sciences, Lund University, Malmö, Sweden (O.M.); Section of Nephrology, Department of Medicine, University of Chicago, Illinois (A.B.C.); Institute of Biomedical Technologies, National Research Centre of Italy, Segrate, Milan, Italy (D.C.); and Sanipedia srl, Bresso, Italy (D.C.)
| | - Anna F Dominiczak
- From the Department of Health Sciences, University of Milan, Italy (E.S., F.R., M.C., M.B., C.B.); Human Genetics and Institute of Molecular Medicine, University of Texas Health Science Center, Houston (Z.W., E.B.); Department of Pharmacotherapy and Translational Research, Center for Pharmacogenomics, College of Pharmacy (Y.G., C.W.M., J.A.J., R.M.C.-D.) and Division of Cardiovascular Medicine, Department of Medicine (J.A.J., R.M.C.-D.), University of Florida, Gainesville; Institute of Cardiovascular and Medical Sciences, College of Medical Veterinary and Life Sciences, University of Glasgow, United Kingdom (S.P., A.F.D.); Department of Medicine, University of Helsinki and Helsinki University Hospital, Finland (T.P.H., K.K.K.); Nephrology and Dialysis and Hypertension Unit, San Raffaele Scientific Institute, Università Vita Salute San Raffaele, Milano, Italy (C.L., P.M.); Hypertension and Related Disease Centre, AOU-University of Sassari, Italy (R.Z., N.G.); Division of Biomedical Statistics and Informatics, Department of Health Sciences Research (K.R.B.) and Division of Nephrology and Hypertension, Department of Internal Medicine (S.T.T.), Mayo Clinic, Rochester, Minnesota; Institute for Molecular Medicine Finland FIMM, University of Helsinki, Finland (A.-P.S); Department of Clinical Sciences, Lund University, Malmö, Sweden (O.M.); Section of Nephrology, Department of Medicine, University of Chicago, Illinois (A.B.C.); Institute of Biomedical Technologies, National Research Centre of Italy, Segrate, Milan, Italy (D.C.); and Sanipedia srl, Bresso, Italy (D.C.)
| | - Julie A Johnson
- From the Department of Health Sciences, University of Milan, Italy (E.S., F.R., M.C., M.B., C.B.); Human Genetics and Institute of Molecular Medicine, University of Texas Health Science Center, Houston (Z.W., E.B.); Department of Pharmacotherapy and Translational Research, Center for Pharmacogenomics, College of Pharmacy (Y.G., C.W.M., J.A.J., R.M.C.-D.) and Division of Cardiovascular Medicine, Department of Medicine (J.A.J., R.M.C.-D.), University of Florida, Gainesville; Institute of Cardiovascular and Medical Sciences, College of Medical Veterinary and Life Sciences, University of Glasgow, United Kingdom (S.P., A.F.D.); Department of Medicine, University of Helsinki and Helsinki University Hospital, Finland (T.P.H., K.K.K.); Nephrology and Dialysis and Hypertension Unit, San Raffaele Scientific Institute, Università Vita Salute San Raffaele, Milano, Italy (C.L., P.M.); Hypertension and Related Disease Centre, AOU-University of Sassari, Italy (R.Z., N.G.); Division of Biomedical Statistics and Informatics, Department of Health Sciences Research (K.R.B.) and Division of Nephrology and Hypertension, Department of Internal Medicine (S.T.T.), Mayo Clinic, Rochester, Minnesota; Institute for Molecular Medicine Finland FIMM, University of Helsinki, Finland (A.-P.S); Department of Clinical Sciences, Lund University, Malmö, Sweden (O.M.); Section of Nephrology, Department of Medicine, University of Chicago, Illinois (A.B.C.); Institute of Biomedical Technologies, National Research Centre of Italy, Segrate, Milan, Italy (D.C.); and Sanipedia srl, Bresso, Italy (D.C.)
| | - Cristina Barlassina
- From the Department of Health Sciences, University of Milan, Italy (E.S., F.R., M.C., M.B., C.B.); Human Genetics and Institute of Molecular Medicine, University of Texas Health Science Center, Houston (Z.W., E.B.); Department of Pharmacotherapy and Translational Research, Center for Pharmacogenomics, College of Pharmacy (Y.G., C.W.M., J.A.J., R.M.C.-D.) and Division of Cardiovascular Medicine, Department of Medicine (J.A.J., R.M.C.-D.), University of Florida, Gainesville; Institute of Cardiovascular and Medical Sciences, College of Medical Veterinary and Life Sciences, University of Glasgow, United Kingdom (S.P., A.F.D.); Department of Medicine, University of Helsinki and Helsinki University Hospital, Finland (T.P.H., K.K.K.); Nephrology and Dialysis and Hypertension Unit, San Raffaele Scientific Institute, Università Vita Salute San Raffaele, Milano, Italy (C.L., P.M.); Hypertension and Related Disease Centre, AOU-University of Sassari, Italy (R.Z., N.G.); Division of Biomedical Statistics and Informatics, Department of Health Sciences Research (K.R.B.) and Division of Nephrology and Hypertension, Department of Internal Medicine (S.T.T.), Mayo Clinic, Rochester, Minnesota; Institute for Molecular Medicine Finland FIMM, University of Helsinki, Finland (A.-P.S); Department of Clinical Sciences, Lund University, Malmö, Sweden (O.M.); Section of Nephrology, Department of Medicine, University of Chicago, Illinois (A.B.C.); Institute of Biomedical Technologies, National Research Centre of Italy, Segrate, Milan, Italy (D.C.); and Sanipedia srl, Bresso, Italy (D.C.)
| | - Eric Boerwinkle
- From the Department of Health Sciences, University of Milan, Italy (E.S., F.R., M.C., M.B., C.B.); Human Genetics and Institute of Molecular Medicine, University of Texas Health Science Center, Houston (Z.W., E.B.); Department of Pharmacotherapy and Translational Research, Center for Pharmacogenomics, College of Pharmacy (Y.G., C.W.M., J.A.J., R.M.C.-D.) and Division of Cardiovascular Medicine, Department of Medicine (J.A.J., R.M.C.-D.), University of Florida, Gainesville; Institute of Cardiovascular and Medical Sciences, College of Medical Veterinary and Life Sciences, University of Glasgow, United Kingdom (S.P., A.F.D.); Department of Medicine, University of Helsinki and Helsinki University Hospital, Finland (T.P.H., K.K.K.); Nephrology and Dialysis and Hypertension Unit, San Raffaele Scientific Institute, Università Vita Salute San Raffaele, Milano, Italy (C.L., P.M.); Hypertension and Related Disease Centre, AOU-University of Sassari, Italy (R.Z., N.G.); Division of Biomedical Statistics and Informatics, Department of Health Sciences Research (K.R.B.) and Division of Nephrology and Hypertension, Department of Internal Medicine (S.T.T.), Mayo Clinic, Rochester, Minnesota; Institute for Molecular Medicine Finland FIMM, University of Helsinki, Finland (A.-P.S); Department of Clinical Sciences, Lund University, Malmö, Sweden (O.M.); Section of Nephrology, Department of Medicine, University of Chicago, Illinois (A.B.C.); Institute of Biomedical Technologies, National Research Centre of Italy, Segrate, Milan, Italy (D.C.); and Sanipedia srl, Bresso, Italy (D.C.)
| | - Rhonda M Cooper-DeHoff
- From the Department of Health Sciences, University of Milan, Italy (E.S., F.R., M.C., M.B., C.B.); Human Genetics and Institute of Molecular Medicine, University of Texas Health Science Center, Houston (Z.W., E.B.); Department of Pharmacotherapy and Translational Research, Center for Pharmacogenomics, College of Pharmacy (Y.G., C.W.M., J.A.J., R.M.C.-D.) and Division of Cardiovascular Medicine, Department of Medicine (J.A.J., R.M.C.-D.), University of Florida, Gainesville; Institute of Cardiovascular and Medical Sciences, College of Medical Veterinary and Life Sciences, University of Glasgow, United Kingdom (S.P., A.F.D.); Department of Medicine, University of Helsinki and Helsinki University Hospital, Finland (T.P.H., K.K.K.); Nephrology and Dialysis and Hypertension Unit, San Raffaele Scientific Institute, Università Vita Salute San Raffaele, Milano, Italy (C.L., P.M.); Hypertension and Related Disease Centre, AOU-University of Sassari, Italy (R.Z., N.G.); Division of Biomedical Statistics and Informatics, Department of Health Sciences Research (K.R.B.) and Division of Nephrology and Hypertension, Department of Internal Medicine (S.T.T.), Mayo Clinic, Rochester, Minnesota; Institute for Molecular Medicine Finland FIMM, University of Helsinki, Finland (A.-P.S); Department of Clinical Sciences, Lund University, Malmö, Sweden (O.M.); Section of Nephrology, Department of Medicine, University of Chicago, Illinois (A.B.C.); Institute of Biomedical Technologies, National Research Centre of Italy, Segrate, Milan, Italy (D.C.); and Sanipedia srl, Bresso, Italy (D.C.)
| | - Stephen T Turner
- From the Department of Health Sciences, University of Milan, Italy (E.S., F.R., M.C., M.B., C.B.); Human Genetics and Institute of Molecular Medicine, University of Texas Health Science Center, Houston (Z.W., E.B.); Department of Pharmacotherapy and Translational Research, Center for Pharmacogenomics, College of Pharmacy (Y.G., C.W.M., J.A.J., R.M.C.-D.) and Division of Cardiovascular Medicine, Department of Medicine (J.A.J., R.M.C.-D.), University of Florida, Gainesville; Institute of Cardiovascular and Medical Sciences, College of Medical Veterinary and Life Sciences, University of Glasgow, United Kingdom (S.P., A.F.D.); Department of Medicine, University of Helsinki and Helsinki University Hospital, Finland (T.P.H., K.K.K.); Nephrology and Dialysis and Hypertension Unit, San Raffaele Scientific Institute, Università Vita Salute San Raffaele, Milano, Italy (C.L., P.M.); Hypertension and Related Disease Centre, AOU-University of Sassari, Italy (R.Z., N.G.); Division of Biomedical Statistics and Informatics, Department of Health Sciences Research (K.R.B.) and Division of Nephrology and Hypertension, Department of Internal Medicine (S.T.T.), Mayo Clinic, Rochester, Minnesota; Institute for Molecular Medicine Finland FIMM, University of Helsinki, Finland (A.-P.S); Department of Clinical Sciences, Lund University, Malmö, Sweden (O.M.); Section of Nephrology, Department of Medicine, University of Chicago, Illinois (A.B.C.); Institute of Biomedical Technologies, National Research Centre of Italy, Segrate, Milan, Italy (D.C.); and Sanipedia srl, Bresso, Italy (D.C.)
| |
Collapse
|
40
|
Abstract
The heritability of hypertension (HTN) is widely recognized and as a result, extensive studies ranging from genetic linkage analyses to genome-wide association studies are actively ongoing to elucidate the etiology of both monogenic and polygenic forms of HTN. Due to the complex nature of essential HTN, however, single genes affecting blood pressure (BP) variability remain difficult to isolate and identify and have rendered the development of single-gene targeted therapies challenging. The roles of other causative factors in modulating BP, such as gene-environment interactions and epigenetic factors, are increasingly being brought to the forefront. In this review, we discuss the various monogenic HTN syndromes and corresponding pathophysiologic mechanisms, the different methodologies employed in genetic studies of essential HTN, the mechanisms for epigenetic modulation of essential HTN, pharmacogenomics and HTN, and finally, recent advances in genetic studies of essential HTN in the pediatric population.
Collapse
Affiliation(s)
- Sun-Young Ahn
- Department of Nephrology, Children's National Health System, Washington, DC, United States.,The George Washington University School of Medicine, Washington, DC, United States
| | - Charu Gupta
- Department of Nephrology, Children's National Health System, Washington, DC, United States.,The George Washington University School of Medicine, Washington, DC, United States
| |
Collapse
|
41
|
Shahin MH, Gong Y, McDonough CW, Rotroff DM, Beitelshees AL, Garrett TJ, Gums JG, Motsinger-Reif A, Chapman AB, Turner ST, Boerwinkle E, Frye RF, Fiehn O, Cooper-DeHoff RM, Kaddurah-Daouk R, Johnson JA. A Genetic Response Score for Hydrochlorothiazide Use: Insights From Genomics and Metabolomics Integration. Hypertension 2016; 68:621-9. [PMID: 27381900 DOI: 10.1161/hypertensionaha.116.07328] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Accepted: 06/05/2016] [Indexed: 12/21/2022]
Abstract
Hydrochlorothiazide is among the most commonly prescribed antihypertensives; yet, <50% of hydrochlorothiazide-treated patients achieve blood pressure (BP) control. Herein, we integrated metabolomic and genomic profiles of hydrochlorothiazide-treated patients to identify novel genetic markers associated with hydrochlorothiazide BP response. The primary analysis included 228 white hypertensives treated with hydrochlorothiazide from the Pharmacogenomic Evaluation of Antihypertensive Responses (PEAR) study. Genome-wide analysis was conducted using Illumina Omni 1 mol/L-Quad Chip, and untargeted metabolomics was performed on baseline fasting plasma samples using a gas chromatography-time-of-flight mass spectrometry platform. We found 13 metabolites significantly associated with hydrochlorothiazide systolic BP (SBP) and diastolic BP (DBP) responses (false discovery rate, <0.05). In addition, integrating genomic and metabolomic data revealed 3 polymorphisms (rs2727563 PRKAG2, rs12604940 DCC, and rs13262930 EPHX2) along with arachidonic acid, converging in the netrin signaling pathway (P=1×10(-5)), as potential markers, significantly influencing hydrochlorothiazide BP response. We successfully replicated the 3 genetic signals in 212 white hypertensives treated with hydrochlorothiazide and created a response score by summing their BP-lowering alleles. We found patients carrying 1 response allele had a significantly lower response than carriers of 6 alleles (∆SBP/∆DBP: -1.5/1.2 versus -16.3/-10.4 mm Hg, respectively, SBP score, P=1×10(-8) and DBP score, P=3×10(-9)). This score explained 11.3% and 11.9% of the variability in hydrochlorothiazide SBP and DBP responses, respectively, and was further validated in another independent study of 196 whites treated with hydrochlorothiazide (DBP score, P=0.03; SBP score, P=0.07). This study suggests that PRKAG2, DCC, and EPHX2 might be important determinants of hydrochlorothiazide BP response.
Collapse
Affiliation(s)
- Mohamed H Shahin
- From the Department of Pharmacotherapy and Translational Research and Center for Pharmacogenomics (M.H.S., Y.G., C.W.M., J.G.G., R.F.F., R.M.C.-D., J.A.J.) and Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine (T.J.G.), University of Florida, Gainesville; Department of Statistics, Bioinformatics Research Center, North Carolina State University, Raleigh (D.M.R., A.M.-R.); Department of Medicine, University of Maryland, Baltimore (A.L.B.); Department of Medicine, Emory University, Atlanta, GA (A.B.C.); Division of Nephrology and Hypertension, Department of Medicine, College of Medicine, Mayo Clinic, Rochester, MN (S.T.T.); Human Genetics Center and Institute of Molecular Medicine, University of Texas Health Science Center, Houston, (E.B.); Department of Molecular and Cellular Biology and Genome Center, University of California, Davis (O.F.); and Department of Psychiatry and Behavioral Sciences, Duke University, Durham, NC (R.K.-D.)
| | - Yan Gong
- From the Department of Pharmacotherapy and Translational Research and Center for Pharmacogenomics (M.H.S., Y.G., C.W.M., J.G.G., R.F.F., R.M.C.-D., J.A.J.) and Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine (T.J.G.), University of Florida, Gainesville; Department of Statistics, Bioinformatics Research Center, North Carolina State University, Raleigh (D.M.R., A.M.-R.); Department of Medicine, University of Maryland, Baltimore (A.L.B.); Department of Medicine, Emory University, Atlanta, GA (A.B.C.); Division of Nephrology and Hypertension, Department of Medicine, College of Medicine, Mayo Clinic, Rochester, MN (S.T.T.); Human Genetics Center and Institute of Molecular Medicine, University of Texas Health Science Center, Houston, (E.B.); Department of Molecular and Cellular Biology and Genome Center, University of California, Davis (O.F.); and Department of Psychiatry and Behavioral Sciences, Duke University, Durham, NC (R.K.-D.)
| | - Caitrin W McDonough
- From the Department of Pharmacotherapy and Translational Research and Center for Pharmacogenomics (M.H.S., Y.G., C.W.M., J.G.G., R.F.F., R.M.C.-D., J.A.J.) and Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine (T.J.G.), University of Florida, Gainesville; Department of Statistics, Bioinformatics Research Center, North Carolina State University, Raleigh (D.M.R., A.M.-R.); Department of Medicine, University of Maryland, Baltimore (A.L.B.); Department of Medicine, Emory University, Atlanta, GA (A.B.C.); Division of Nephrology and Hypertension, Department of Medicine, College of Medicine, Mayo Clinic, Rochester, MN (S.T.T.); Human Genetics Center and Institute of Molecular Medicine, University of Texas Health Science Center, Houston, (E.B.); Department of Molecular and Cellular Biology and Genome Center, University of California, Davis (O.F.); and Department of Psychiatry and Behavioral Sciences, Duke University, Durham, NC (R.K.-D.)
| | - Daniel M Rotroff
- From the Department of Pharmacotherapy and Translational Research and Center for Pharmacogenomics (M.H.S., Y.G., C.W.M., J.G.G., R.F.F., R.M.C.-D., J.A.J.) and Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine (T.J.G.), University of Florida, Gainesville; Department of Statistics, Bioinformatics Research Center, North Carolina State University, Raleigh (D.M.R., A.M.-R.); Department of Medicine, University of Maryland, Baltimore (A.L.B.); Department of Medicine, Emory University, Atlanta, GA (A.B.C.); Division of Nephrology and Hypertension, Department of Medicine, College of Medicine, Mayo Clinic, Rochester, MN (S.T.T.); Human Genetics Center and Institute of Molecular Medicine, University of Texas Health Science Center, Houston, (E.B.); Department of Molecular and Cellular Biology and Genome Center, University of California, Davis (O.F.); and Department of Psychiatry and Behavioral Sciences, Duke University, Durham, NC (R.K.-D.)
| | - Amber L Beitelshees
- From the Department of Pharmacotherapy and Translational Research and Center for Pharmacogenomics (M.H.S., Y.G., C.W.M., J.G.G., R.F.F., R.M.C.-D., J.A.J.) and Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine (T.J.G.), University of Florida, Gainesville; Department of Statistics, Bioinformatics Research Center, North Carolina State University, Raleigh (D.M.R., A.M.-R.); Department of Medicine, University of Maryland, Baltimore (A.L.B.); Department of Medicine, Emory University, Atlanta, GA (A.B.C.); Division of Nephrology and Hypertension, Department of Medicine, College of Medicine, Mayo Clinic, Rochester, MN (S.T.T.); Human Genetics Center and Institute of Molecular Medicine, University of Texas Health Science Center, Houston, (E.B.); Department of Molecular and Cellular Biology and Genome Center, University of California, Davis (O.F.); and Department of Psychiatry and Behavioral Sciences, Duke University, Durham, NC (R.K.-D.)
| | - Timothy J Garrett
- From the Department of Pharmacotherapy and Translational Research and Center for Pharmacogenomics (M.H.S., Y.G., C.W.M., J.G.G., R.F.F., R.M.C.-D., J.A.J.) and Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine (T.J.G.), University of Florida, Gainesville; Department of Statistics, Bioinformatics Research Center, North Carolina State University, Raleigh (D.M.R., A.M.-R.); Department of Medicine, University of Maryland, Baltimore (A.L.B.); Department of Medicine, Emory University, Atlanta, GA (A.B.C.); Division of Nephrology and Hypertension, Department of Medicine, College of Medicine, Mayo Clinic, Rochester, MN (S.T.T.); Human Genetics Center and Institute of Molecular Medicine, University of Texas Health Science Center, Houston, (E.B.); Department of Molecular and Cellular Biology and Genome Center, University of California, Davis (O.F.); and Department of Psychiatry and Behavioral Sciences, Duke University, Durham, NC (R.K.-D.)
| | - John G Gums
- From the Department of Pharmacotherapy and Translational Research and Center for Pharmacogenomics (M.H.S., Y.G., C.W.M., J.G.G., R.F.F., R.M.C.-D., J.A.J.) and Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine (T.J.G.), University of Florida, Gainesville; Department of Statistics, Bioinformatics Research Center, North Carolina State University, Raleigh (D.M.R., A.M.-R.); Department of Medicine, University of Maryland, Baltimore (A.L.B.); Department of Medicine, Emory University, Atlanta, GA (A.B.C.); Division of Nephrology and Hypertension, Department of Medicine, College of Medicine, Mayo Clinic, Rochester, MN (S.T.T.); Human Genetics Center and Institute of Molecular Medicine, University of Texas Health Science Center, Houston, (E.B.); Department of Molecular and Cellular Biology and Genome Center, University of California, Davis (O.F.); and Department of Psychiatry and Behavioral Sciences, Duke University, Durham, NC (R.K.-D.)
| | - Alison Motsinger-Reif
- From the Department of Pharmacotherapy and Translational Research and Center for Pharmacogenomics (M.H.S., Y.G., C.W.M., J.G.G., R.F.F., R.M.C.-D., J.A.J.) and Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine (T.J.G.), University of Florida, Gainesville; Department of Statistics, Bioinformatics Research Center, North Carolina State University, Raleigh (D.M.R., A.M.-R.); Department of Medicine, University of Maryland, Baltimore (A.L.B.); Department of Medicine, Emory University, Atlanta, GA (A.B.C.); Division of Nephrology and Hypertension, Department of Medicine, College of Medicine, Mayo Clinic, Rochester, MN (S.T.T.); Human Genetics Center and Institute of Molecular Medicine, University of Texas Health Science Center, Houston, (E.B.); Department of Molecular and Cellular Biology and Genome Center, University of California, Davis (O.F.); and Department of Psychiatry and Behavioral Sciences, Duke University, Durham, NC (R.K.-D.)
| | - Arlene B Chapman
- From the Department of Pharmacotherapy and Translational Research and Center for Pharmacogenomics (M.H.S., Y.G., C.W.M., J.G.G., R.F.F., R.M.C.-D., J.A.J.) and Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine (T.J.G.), University of Florida, Gainesville; Department of Statistics, Bioinformatics Research Center, North Carolina State University, Raleigh (D.M.R., A.M.-R.); Department of Medicine, University of Maryland, Baltimore (A.L.B.); Department of Medicine, Emory University, Atlanta, GA (A.B.C.); Division of Nephrology and Hypertension, Department of Medicine, College of Medicine, Mayo Clinic, Rochester, MN (S.T.T.); Human Genetics Center and Institute of Molecular Medicine, University of Texas Health Science Center, Houston, (E.B.); Department of Molecular and Cellular Biology and Genome Center, University of California, Davis (O.F.); and Department of Psychiatry and Behavioral Sciences, Duke University, Durham, NC (R.K.-D.)
| | - Stephen T Turner
- From the Department of Pharmacotherapy and Translational Research and Center for Pharmacogenomics (M.H.S., Y.G., C.W.M., J.G.G., R.F.F., R.M.C.-D., J.A.J.) and Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine (T.J.G.), University of Florida, Gainesville; Department of Statistics, Bioinformatics Research Center, North Carolina State University, Raleigh (D.M.R., A.M.-R.); Department of Medicine, University of Maryland, Baltimore (A.L.B.); Department of Medicine, Emory University, Atlanta, GA (A.B.C.); Division of Nephrology and Hypertension, Department of Medicine, College of Medicine, Mayo Clinic, Rochester, MN (S.T.T.); Human Genetics Center and Institute of Molecular Medicine, University of Texas Health Science Center, Houston, (E.B.); Department of Molecular and Cellular Biology and Genome Center, University of California, Davis (O.F.); and Department of Psychiatry and Behavioral Sciences, Duke University, Durham, NC (R.K.-D.)
| | - Eric Boerwinkle
- From the Department of Pharmacotherapy and Translational Research and Center for Pharmacogenomics (M.H.S., Y.G., C.W.M., J.G.G., R.F.F., R.M.C.-D., J.A.J.) and Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine (T.J.G.), University of Florida, Gainesville; Department of Statistics, Bioinformatics Research Center, North Carolina State University, Raleigh (D.M.R., A.M.-R.); Department of Medicine, University of Maryland, Baltimore (A.L.B.); Department of Medicine, Emory University, Atlanta, GA (A.B.C.); Division of Nephrology and Hypertension, Department of Medicine, College of Medicine, Mayo Clinic, Rochester, MN (S.T.T.); Human Genetics Center and Institute of Molecular Medicine, University of Texas Health Science Center, Houston, (E.B.); Department of Molecular and Cellular Biology and Genome Center, University of California, Davis (O.F.); and Department of Psychiatry and Behavioral Sciences, Duke University, Durham, NC (R.K.-D.)
| | - Reginald F Frye
- From the Department of Pharmacotherapy and Translational Research and Center for Pharmacogenomics (M.H.S., Y.G., C.W.M., J.G.G., R.F.F., R.M.C.-D., J.A.J.) and Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine (T.J.G.), University of Florida, Gainesville; Department of Statistics, Bioinformatics Research Center, North Carolina State University, Raleigh (D.M.R., A.M.-R.); Department of Medicine, University of Maryland, Baltimore (A.L.B.); Department of Medicine, Emory University, Atlanta, GA (A.B.C.); Division of Nephrology and Hypertension, Department of Medicine, College of Medicine, Mayo Clinic, Rochester, MN (S.T.T.); Human Genetics Center and Institute of Molecular Medicine, University of Texas Health Science Center, Houston, (E.B.); Department of Molecular and Cellular Biology and Genome Center, University of California, Davis (O.F.); and Department of Psychiatry and Behavioral Sciences, Duke University, Durham, NC (R.K.-D.)
| | - Oliver Fiehn
- From the Department of Pharmacotherapy and Translational Research and Center for Pharmacogenomics (M.H.S., Y.G., C.W.M., J.G.G., R.F.F., R.M.C.-D., J.A.J.) and Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine (T.J.G.), University of Florida, Gainesville; Department of Statistics, Bioinformatics Research Center, North Carolina State University, Raleigh (D.M.R., A.M.-R.); Department of Medicine, University of Maryland, Baltimore (A.L.B.); Department of Medicine, Emory University, Atlanta, GA (A.B.C.); Division of Nephrology and Hypertension, Department of Medicine, College of Medicine, Mayo Clinic, Rochester, MN (S.T.T.); Human Genetics Center and Institute of Molecular Medicine, University of Texas Health Science Center, Houston, (E.B.); Department of Molecular and Cellular Biology and Genome Center, University of California, Davis (O.F.); and Department of Psychiatry and Behavioral Sciences, Duke University, Durham, NC (R.K.-D.)
| | - Rhonda M Cooper-DeHoff
- From the Department of Pharmacotherapy and Translational Research and Center for Pharmacogenomics (M.H.S., Y.G., C.W.M., J.G.G., R.F.F., R.M.C.-D., J.A.J.) and Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine (T.J.G.), University of Florida, Gainesville; Department of Statistics, Bioinformatics Research Center, North Carolina State University, Raleigh (D.M.R., A.M.-R.); Department of Medicine, University of Maryland, Baltimore (A.L.B.); Department of Medicine, Emory University, Atlanta, GA (A.B.C.); Division of Nephrology and Hypertension, Department of Medicine, College of Medicine, Mayo Clinic, Rochester, MN (S.T.T.); Human Genetics Center and Institute of Molecular Medicine, University of Texas Health Science Center, Houston, (E.B.); Department of Molecular and Cellular Biology and Genome Center, University of California, Davis (O.F.); and Department of Psychiatry and Behavioral Sciences, Duke University, Durham, NC (R.K.-D.)
| | - Rima Kaddurah-Daouk
- From the Department of Pharmacotherapy and Translational Research and Center for Pharmacogenomics (M.H.S., Y.G., C.W.M., J.G.G., R.F.F., R.M.C.-D., J.A.J.) and Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine (T.J.G.), University of Florida, Gainesville; Department of Statistics, Bioinformatics Research Center, North Carolina State University, Raleigh (D.M.R., A.M.-R.); Department of Medicine, University of Maryland, Baltimore (A.L.B.); Department of Medicine, Emory University, Atlanta, GA (A.B.C.); Division of Nephrology and Hypertension, Department of Medicine, College of Medicine, Mayo Clinic, Rochester, MN (S.T.T.); Human Genetics Center and Institute of Molecular Medicine, University of Texas Health Science Center, Houston, (E.B.); Department of Molecular and Cellular Biology and Genome Center, University of California, Davis (O.F.); and Department of Psychiatry and Behavioral Sciences, Duke University, Durham, NC (R.K.-D.)
| | - Julie A Johnson
- From the Department of Pharmacotherapy and Translational Research and Center for Pharmacogenomics (M.H.S., Y.G., C.W.M., J.G.G., R.F.F., R.M.C.-D., J.A.J.) and Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine (T.J.G.), University of Florida, Gainesville; Department of Statistics, Bioinformatics Research Center, North Carolina State University, Raleigh (D.M.R., A.M.-R.); Department of Medicine, University of Maryland, Baltimore (A.L.B.); Department of Medicine, Emory University, Atlanta, GA (A.B.C.); Division of Nephrology and Hypertension, Department of Medicine, College of Medicine, Mayo Clinic, Rochester, MN (S.T.T.); Human Genetics Center and Institute of Molecular Medicine, University of Texas Health Science Center, Houston, (E.B.); Department of Molecular and Cellular Biology and Genome Center, University of California, Davis (O.F.); and Department of Psychiatry and Behavioral Sciences, Duke University, Durham, NC (R.K.-D.).
| |
Collapse
|
42
|
Byrd JB. Personalized medicine and treatment approaches in hypertension: current perspectives. Integr Blood Press Control 2016; 9:59-67. [PMID: 27103841 PMCID: PMC4827884 DOI: 10.2147/ibpc.s74320] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
In the US, hypertension affects one in three adults. Current guideline-based treatment of hypertension involves little diagnostic testing. A more personalized approach to the treatment of hypertension might be of use. Several methods of personalized treatment have been proposed and vetted to varying degrees. The purpose of this narrative review is to discuss the rationale for personalized therapy in hypertension, barriers to its development and implementation, some influential examples of proposed personalization measures, and a view of future efforts.
Collapse
Affiliation(s)
- James Brian Byrd
- Division of Cardiovascular Medicine, Department of Medicine, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
43
|
TET2 and CSMD1 genes affect SBP response to hydrochlorothiazide in never-treated essential hypertensives. J Hypertens 2016; 33:1301-9. [PMID: 25695618 DOI: 10.1097/hjh.0000000000000541] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Thiazide diuretics have been recommended as a first-line antihypertensive treatment, although the choice of 'the right drug in the individual essential hypertensive patient' remains still empirical. Essential hypertension is a complex, polygenic disease derived from the interaction of patient's genetic background with the environment. Pharmacogenomics could be a useful tool to pinpoint gene variants involved in antihypertensive drug response, thus optimizing therapeutic advantages and minimizing side effects. METHODS AND RESULTS We looked for variants associated with blood pressure response to hydrochlorothiazide over an 8-week follow-up by means of a genome-wide association analysis in two Italian cohorts of never-treated essential hypertensive patients: 343 samples from Sardinia and 142 from Milan. TET2 and CSMD1 as plausible candidate genes to affect SBP response to hydrochlorothiazide were identified. The specificity of our findings for hydrochlorothiazide was confirmed in an independent cohort of essential hypertensive patients treated with losartan. Our best findings were also tested for replication in four independent hypertensive samples of European Ancestry, such as GENetics of drug RESponsiveness in essential hypertension, Genetic Epidemiology of Responses to Antihypertensives, NORdic DILtiazem intervention, Pharmacogenomics Evaluation of Antihypertensive Responses, and Campania Salute Network-StayOnDiur. We validated a polymorphism in CSMD1 and UGGT2. CONCLUSION This exploratory study reports two plausible loci associated with SBP response to hydrochlorothiazide: TET2, an aldosterone-responsive mediator of αENaC gene transcription; and CSMD1, previously described as associated with hypertension in a case-control study.
Collapse
|
44
|
Eadon MT, Chapman AB. A Physiologic Approach to the Pharmacogenomics of Hypertension. Adv Chronic Kidney Dis 2016; 23:91-105. [PMID: 26979148 DOI: 10.1053/j.ackd.2016.02.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Hypertension is a multifactorial condition with diverse physiological systems contributing to its pathogenesis. Individuals exhibit significant variation in their response to antihypertensive agents. Traditional markers, such as age, gender, diet, plasma renin level, and ethnicity, aid in drug selection. However, this review explores the contribution of genetics to facilitate antihypertensive agent selection and predict treatment efficacy. The findings, reproducibility, and limitations of published studies are examined, with emphasis placed on candidate genetic variants affecting drug metabolism, the renin-angiotensin system, adrenergic signalling, and renal sodium reabsorption. Single-nucleotide polymorphisms identified and replicated in unbiased genome-wide association studies of hypertension treatment are reviewed to illustrate the evolving understanding of the disease's complex and polygenic pathophysiology. Implementation efforts at academic centers seek to overcome barriers to the broad adoption of pharmacogenomics in the treatment of hypertension. The level of evidence required to support the implementation of pharmacogenomics in clinical practice is considered.
Collapse
|
45
|
Shahin MH, Johnson JA. Mechanisms and pharmacogenetic signals underlying thiazide diuretics blood pressure response. Curr Opin Pharmacol 2016; 27:31-7. [PMID: 26874237 DOI: 10.1016/j.coph.2016.01.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Accepted: 01/12/2016] [Indexed: 01/13/2023]
Abstract
Thiazide (TZD) diuretics are among the most commonly prescribed antihypertensives globally; however their chronic blood pressure (BP) lowering mechanism remains unclear. Herein we discuss the current evidence regarding specific mechanisms regulating the antihypertensive effects of TZDs, suggesting that TZDs act via multiple complex and interacting mechanisms, including natriuresis with short term use and direct vasodilatory effects chronically. Additionally, we review pharmacogenomics signals that have been associated with TZDs BP-response in several cohorts (i.e. NEDD4L, PRKCA, EDNRA-GNAS, and YEATS4) and discuss how these genes might be related to TZD BP-response mechanism. Understanding the association between these genes and TZD BP mechanism might facilitate the development of new drugs and therapeutic approaches based on a deeper understanding of the determinants of BP-response.
Collapse
Affiliation(s)
- Mohamed H Shahin
- Department of Pharmacotherapy and Translational Research, Center for Pharmacogenomics, College of Pharmacy, University of Florida, Gainesville, FL, USA
| | - Julie A Johnson
- Department of Pharmacotherapy and Translational Research, Center for Pharmacogenomics, College of Pharmacy, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
46
|
Cooper-DeHoff RM, Johnson JA. Hypertension pharmacogenomics: in search of personalized treatment approaches. Nat Rev Nephrol 2016; 12:110-22. [PMID: 26592190 PMCID: PMC4778736 DOI: 10.1038/nrneph.2015.176] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Cardiovascular and renal diseases are associated with many risk factors, of which hypertension is one of the most prevalent. Worldwide, blood pressure control is only achieved in ∼50% of those treated for hypertension, despite the availability of a considerable number of antihypertensive drugs from different pharmacological classes. Although many reasons exist for poor blood pressure control, a likely contributor is the inability to predict to which antihypertensive drug an individual is most likely to respond. Hypertension pharmacogenomics and other 'omics' technologies have the potential to identify genetic signals that are predictive of response or adverse outcome to particular drugs, and guide selection of hypertension treatment for a given individual. Continued research in this field will enhance our understanding of how to maximally deploy the various antihypertensive drug classes to optimize blood pressure response at the individual level. This Review summarizes the available literature on the most convincing genetic signals associated with antihypertensive drug responses and adverse cardiovascular outcomes. Future research in this area will be facilitated by enhancing collaboration between research groups through consortia such as the International Consortium for Antihypertensives Pharmacogenomics Studies, with the goal of translating replicated findings into clinical implementation.
Collapse
Affiliation(s)
- Rhonda M Cooper-DeHoff
- Department of Pharmacotherapy and Translational Research and Division of Cardiovascular Medicine, Colleges of Pharmacy and Medicine, University of Florida, PO Box 100484, 1600 SW Archer Road, Gainesville, Florida 32610-0484, USA
| | - Julie A Johnson
- Department of Pharmacotherapy and Translational Research and Division of Cardiovascular Medicine, Colleges of Pharmacy and Medicine, University of Florida, PO Box 100484, 1600 SW Archer Road, Gainesville, Florida 32610-0484, USA
| |
Collapse
|
47
|
The effects of genes implicated in cardiovascular disease on blood pressure response to treatment among treatment-naive hypertensive African Americans in the GenHAT study. J Hum Hypertens 2016; 30:549-54. [PMID: 26791477 PMCID: PMC4956602 DOI: 10.1038/jhh.2015.121] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Revised: 09/29/2015] [Accepted: 10/19/2015] [Indexed: 12/24/2022]
Abstract
African Americans have the highest prevalence of hypertension in the United States. Blood-pressure control is important to reduce cardiovascular disease (CVD)-related morbidity and mortality in this ethnic group. Genetic variants have been found to be associated with BP response to treatment. Previous pharmacogenetic studies of blood-pressure response to treatment in African Americans suffer limitations of small sample size as well as a limited number of candidate genes, and often focused on one antihypertensive treatment. Using 1,131 African-American treatment naïve participants from the Genetics of Hypertension Associated Treatment (GenHAT) Study, we examined whether variants in 35 candidate genes might modulate blood-pressure response to four different antihypertensive medications, including an angiotensin converting enzyme (ACE) inhibitor (lisinopril), a calcium channel blocker (amlodipine), and an α-adrenergic blocker (doxazosin) as compared to a thiazide diuretic (chlorthalidone) after 6 months of follow-up. Several suggestive gene by treatment interactions were identified. For example, among participants with two minor alleles of REN rs6681776, diastolic blood-pressure response was much improved on doxazosin compared to chlorthalidone (on average −9.49 mmHg vs. −1.70 mmHg) (P=0.007). Although several suggestive loci were identified, none of the findings passed significance criteria after correction for multiple testing. Given the impact of hypertension and its sequelae in this population, this research highlights the potential for genetic factors to contribute to blood-pressure response to treatment. Continued concerted research efforts focused on genetics are needed to improve treatment response in this high risk group.
Collapse
|
48
|
Taylor JY, Schwander K, Kardia SLR, Arnett D, Liang J, Hunt SC, Rao DC, Sun YV. A Genome-wide study of blood pressure in African Americans accounting for gene-smoking interaction. Sci Rep 2016; 6:18812. [PMID: 26752167 PMCID: PMC4707536 DOI: 10.1038/srep18812] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2015] [Accepted: 11/09/2015] [Indexed: 12/28/2022] Open
Abstract
Cigarette smoking has been shown to be a health hazard. In addition to being considered a negative lifestyle behavior, studies have shown that cigarette smoking has been linked to genetic underpinnings of hypertension. Because African Americans have the highest incidence and prevalence of hypertension, we examined the joint effect of genetics and cigarette smoking on health among this understudied population. The sample included African Americans from the genome wide association studies of HyperGEN (N = 1083, discovery sample) and GENOA (N = 1427, replication sample), both part of the FBPP. Results suggested that 2 SNPs located on chromosomes 14 (NEDD8; rs11158609; raw p = 9.80 × 10−9, genomic control-adjusted p = 2.09 × 10−7) and 17 (TTYH2; rs8078051; raw p = 6.28 × 10−8, genomic control-adjusted p = 9.65 × 10−7) were associated with SBP including the genetic interaction with cigarette smoking. These two SNPs were not associated with SBP in a main genetic effect only model. This study advances knowledge in the area of main and joint effects of genetics and cigarette smoking on hypertension among African Americans and offers a model to the reader for assessing these risks. More research is required to determine how these genes play a role in expression of hypertension.
Collapse
Affiliation(s)
| | - Karen Schwander
- Division of Biostatistics, School of Medicine, Washington University in St. Louis, St. Louis
| | - Sharon L R Kardia
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor
| | - Donna Arnett
- Department of Epidemiology, School of Public Health, University of Alabama, Birmingham
| | - Jingjing Liang
- Department of Epidemiology and Biostatistics, School of Medicine, Case Western Reserve University, Cleveland
| | - Steven C Hunt
- Cardiovascular Genetics Division, School of Medicine, University of Utah, Salt Lake City
| | - D C Rao
- Division of Biostatistics, School of Medicine, Washington University in St. Louis, St. Louis
| | - Yan V Sun
- Department of Epidemiology, Rollins School of Public Health, Emory University.,Department of Biomedical Informatics, School of Medicine, Emory University, Atlanta
| |
Collapse
|
49
|
Abstract
Heart disease is a leading cause of death in the United States, and hypertension is a predominant risk factor. Thus, effective blood pressure control is important to prevent adverse sequelae of hypertension, including heart failure, coronary artery disease, atrial fibrillation, and ischemic stroke. Over half of Americans have uncontrolled blood pressure, which may in part be explained by interpatient variability in drug response secondary to genetic polymorphism. As such, pharmacogenetic testing may be a supplementary tool to guide treatment. This review highlights the pharmacogenetics of antihypertensive response and response to drugs that treat adverse hypertension-related sequelae, particularly coronary artery disease and atrial fibrillation. While pharmacogenetic evidence may be more robust for the latter with respect to clinical implementation, there is increasing evidence of genetic variants that may help predict antihypertensive response. However, additional research and validation are needed before clinical implementation guidelines for antihypertensive therapy can become a reality.
Collapse
|
50
|
Lupoli S, Salvi E, Barcella M, Barlassina C. Pharmacogenomics considerations in the control of hypertension. Pharmacogenomics 2015; 16:1951-64. [PMID: 26555875 DOI: 10.2217/pgs.15.131] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The response to antihypertensive therapy is very heterogeneous and the need by the physicians to account for it has driven much interest in pharmacogenomics of antihypertensive drugs. The Human Genome Project and the initiatives in genomics that followed, generated a huge number of genetic data that furnished the tools to explore the genotype-phenotype association in candidate genes and at genome-wide level. In spite of the efforts and the great number of publications, pharmacogenomics of antihypertensive drugs is far from being used in clinical practice. In this review, we analyze the main findings available in PubMed from 2010 to 2015, in relation to the major classes of antihypertensive drugs. We also describe a new Phase II drug that targets two specific hypertension predisposing mechanisms.
Collapse
Affiliation(s)
- Sara Lupoli
- Department of Health Sciences, Milan University, Via Rudinì 8, 20142 Milan & Filarete Foundation, Viale Ortles 22/4, 20139 Milan, Italy
| | - Erika Salvi
- Department of Health Sciences, Milan University, Via Rudinì 8, 20142 Milan & Filarete Foundation, Viale Ortles 22/4, 20139 Milan, Italy
| | - Matteo Barcella
- Department of Health Sciences, Milan University, Via Rudinì 8, 20142 Milan & Filarete Foundation, Viale Ortles 22/4, 20139 Milan, Italy
| | - Cristina Barlassina
- Department of Health Sciences, Milan University, Via Rudinì 8, 20142 Milan & Filarete Foundation, Viale Ortles 22/4, 20139 Milan, Italy
| |
Collapse
|